201
|
Zhou S, Zhang H. Synergies of Targeting Angiogenesis and Immune Checkpoints in Cancer: From Mechanism to Clinical Applications. Anticancer Agents Med Chem 2021; 20:768-776. [PMID: 32031076 DOI: 10.2174/1871520620666200207091653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/23/2019] [Accepted: 01/12/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Angiogenesis marks key progress in the growth, recurrence, and metastasis of various cancers. Antiangiogenic drugs can improve the blood supply and oxygen content of tumors and enhance the effects of chemotherapy and radiotherapy by normalizing tumor blood vessels and microenvironment. The further recent developments of Immune Checkpoint Inhibitors (ICIs) provide significant progress in cancer immunotherapy. The study focused on programmed cell death protein 1 (PD-1) and Cytotoxic T Lymphocyte Antigen 4 (CTLA-4) blockade, reflecting on the evidence of durable responses among various tumor types. The aim of this review was to sum up present evidence and clarify the rationale behind supporting the benefits of combining antiangiogenic drugs with immunotherapy for cancer treatment as well as list the ongoing clinical trials that are being conducted. METHODS Using PubMed and Web of Science, published articles have been searched and comprehensively reviewed. RESULTS Antiangiogenic agents can trigger antitumor and immunity, and they can also be induced by the immune system. Combining antiangiogenic drugs with immunotherapy may be effective for the treatment of human cancers. CONCLUSION It is evidenced that combining angiogenesis inhibitors with immunotherapy has a synergistic effect thus improving the curative effect of both agents.
Collapse
Affiliation(s)
- Shi Zhou
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, Jiangsu, China
| | - Haijun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
202
|
de Miranda FS, Guimarães JPT, Menikdiwela KR, Mabry B, Dhakal R, Rahman RL, Moussa H, Moustaid-Moussa N. Breast cancer and the renin-angiotensin system (RAS): Therapeutic approaches and related metabolic diseases. Mol Cell Endocrinol 2021; 528:111245. [PMID: 33753205 DOI: 10.1016/j.mce.2021.111245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
The Renin-Angiotensin System (RAS) is classically recognized for regulating blood pressure and fluid balance. Recently, this role has extended to other areas including inflammation, obesity, diabetes, as well as breast cancer. RAS components are expressed in normal and cancerous breast tissues, and downregulation of RAS inhibits metastasis, proliferation, angiogenesis, and desmoplasia in the tumor microenvironment. Therefore, RAS inhibitors (Angiotensin receptor blockers, ARBs, or angiotensin converting enzyme inhibitors, ACE-I) may be beneficial as preventive adjuvant therapies to thwart breast cancer development and improve outcomes, respectively. Given the beneficial effects of RAS inhibitors in metabolic diseases, which often co-exist in breast cancer patients, combining RAS inhibitors with other breast cancer therapies may enhance the effectiveness of current treatments. This review scrutinizes above associations, to advance our understanding of the role of RAS in breast cancer and its potential for repurposing of RAS inhibitors to improve the therapeutic approach for breast cancer patients.
Collapse
Affiliation(s)
- Flávia Sardela de Miranda
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - João Pedro Tôrres Guimarães
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA; Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo (ICB/USP), São Paulo, SP, Brazil; Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo (FCF/USP), São Paulo, SP, Brazil
| | - Kalhara R Menikdiwela
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Brennan Mabry
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA
| | - Rabin Dhakal
- Department of Mechanical Engineering, Texas Tech University (TTU), Lubbock, TX, USA
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hanna Moussa
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA; Department of Mechanical Engineering, Texas Tech University (TTU), Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Laboratory of Nutrigenomics, Inflammation and Obesity Research, Department of Nutritional Sciences, Texas Tech University (TTU), Lubbock, TX, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
203
|
Tan Y, Chen Q, Li X, Zeng Z, Xiong W, Li G, Li X, Yang J, Xiang B, Yi M. Pyroptosis: a new paradigm of cell death for fighting against cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:153. [PMID: 33941231 PMCID: PMC8091792 DOI: 10.1186/s13046-021-01959-x] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Unraveling the mystery of cell death is one of the most fundamental progresses of life sciences during the past decades. Regulated cell death (RCD) or programmed cell death (PCD) is not only essential in embryonic development, but also plays an important role in the occurrence and progression of diseases, especially cancers. Escaping of cell death is one of hallmarks of cancer. MAIN BODY Pyroptosis is an inflammatory cell death usually caused by microbial infection, accompanied by activation of inflammasomes and maturation of pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18). Gasdermin family proteins are the executors of pyroptosis. Cytotoxic N-terminal of gasdermins generated from caspases or granzymes proteases mediated cleavage of gasdermin proteins oligomerizes and forms pore across cell membrane, leading to release of IL-1β, IL-18. Pyroptosis exerts tumor suppression function and evokes anti-tumor immune responses. Therapeutic regimens, including chemotherapy, radiotherapy, targeted therapy and immune therapy, induce pyroptosis in cancer, which potentiate local and systemic anti-tumor immunity. On the other hand, pyroptosis of normal cells attributes to side effects of anti-cancer therapies. CONCLUSION In this review, we focus on the regulatory mechanisms of pyroptosis and the tumor suppressive function of pyroptosis. We discuss the attribution of pyroptosis in reprogramming tumor microenvironments and restoration of anti-tumor immunity and its potential application in cancer immune therapy.
Collapse
Affiliation(s)
- Yixin Tan
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.,Department of Dermatology, The Second Xiangya Hospital, The Central South University, Changsha, 410011, Hunan, China
| | - Quanzhu Chen
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Jianbo Yang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Mei Yi
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China. .,Department of Dermatology, Xiangya Hospital, The Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
204
|
Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers (Basel) 2021; 13:cancers13092053. [PMID: 33922795 PMCID: PMC8122975 DOI: 10.3390/cancers13092053] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Metastasis, the process by which cancer cells escape primary tumor site and colonize distant organs, is responsible for most cancer-related deaths. The tumor microenvironment (TME), comprises different cell types, including immune cells and cancer-associated fibroblasts, as well as structural elements, such as collagen and hyaluronan that constitute the extracellular matrix (ECM). Intratumoral interactions between the cellular and structural components of the TME regulate the aggressiveness, and dissemination of malignant cells and promote immune evasion. At the secondary site, the TME also facilitates escape from dormancy to enhance metastatic tumor outgrowth. Moreover, the ECM applies mechanical forces on tumors that contribute to hypoxia and cancer cell invasiveness whereas also hinders drug delivery and efficacy in both primary and metastatic sites. In this review, we summarize the latest developments regarding the role of the TME in cancer progression and discuss ongoing efforts to remodel the TME to stop metastasis in its tracks. Abstract The tumor microenvironment (TME) regulates essential tumor survival and promotion functions. Interactions between the cellular and structural components of the TME allow cancer cells to become invasive and disseminate from the primary site to distant locations, through a complex and multistep metastatic cascade. Tumor-associated M2-type macrophages have growth-promoting and immunosuppressive functions; mesenchymal cells mass produce exosomes that increase the migratory ability of cancer cells; cancer associated fibroblasts (CAFs) reorganize the surrounding matrix creating migration-guiding tracks for cancer cells. In addition, the tumor extracellular matrix (ECM) exerts determinant roles in disease progression and cancer cell migration and regulates therapeutic responses. The hypoxic conditions generated at the primary tumor force cancer cells to genetically and/or epigenetically adapt in order to survive and metastasize. In the circulation, cancer cells encounter platelets, immune cells, and cytokines in the blood microenvironment that facilitate their survival and transit. This review discusses the roles of different cellular and structural tumor components in regulating the metastatic process, targeting approaches using small molecule inhibitors, nanoparticles, manipulated exosomes, and miRNAs to inhibit tumor invasion as well as current and future strategies to remodel the TME and enhance treatment efficacy to block the detrimental process of metastasis.
Collapse
Affiliation(s)
- Christiana M. Neophytou
- European University Research Center, Nicosia 2404, Cyprus;
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 1516, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.P.); (T.S.)
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.P.); (T.S.)
| | - Panagiotis Papageorgis
- European University Research Center, Nicosia 2404, Cyprus;
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 1516, Cyprus
- Correspondence: ; Tel.: +357-22-713158
| |
Collapse
|
205
|
Zhang JY, Xue WJ, Wang M, Li W, Dong R, Li MT, Sun LP. Discovery of 4,6-Disubstituted Pyrimidine Derivatives as Novel Dual VEGFR2/FGFR1 Inhibitors. Chem Biodivers 2021; 18:e2100095. [PMID: 33829649 DOI: 10.1002/cbdv.202100095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/06/2021] [Indexed: 11/08/2022]
Abstract
Abnormalities in the FGFRs signaling pathway and VEGFR2 amplification often occur in a variety of tumors, and they synergistically promote tumor angiogenesis. Studies have shown that the up-regulation of FGF-2 is closely related to the resistance of VEGFR2 inhibitors. Activation of the FGFRs signal is a signal of compensatory angiogenesis after VEGFR2 resistance. Dual VEGFR2/FGFR1 inhibitors contribute to overcoming the resistance of VEGFR2 inhibitors and inhibit tumor growth significantly. Based on this, we designed and synthesized a series of 4,6-disubstituted pyrimidine derivatives as dual VEGFR2/FGFR1 inhibitors by the molecular hybridization strategy. 3-(2,6-Dichloro-3,5-dimethoxyphenyl)-1-{6-[(4-methoxyphenyl)amino]pyrimidin-4-yl}-1-methylurea (8b) had the best inhibitory activities against VEGFR2 and FGFR1 at 10 μM (82.2 % and 101.0 %, respectively), it showed moderate antiproliferative activities against A549 and KG-1 cell lines as well. Besides, molecular docking was also carried out to study the binding mode of 3-(2,6-dichloro-3,5-dimethoxyphenyl)-1-{6-[(4-methoxyphenyl)-amino]-pyrimidin-4-yl}-1-methylurea (8b) with VEGFR2 and FGFR1. These studies reveal that this series of compounds deserve further optimization.
Collapse
Affiliation(s)
- Jin-Yang Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Wen-Jun Xue
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Min Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Wen Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Ru Dong
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Ming-Tao Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Li-Ping Sun
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| |
Collapse
|
206
|
Martín-Beltrán C, Gil-Edo R, Hernández-Ribelles G, Agut R, Marí-Mezquita P, Carda M, Falomir E. Aryl Urea Based Scaffolds for Multitarget Drug Discovery in Anticancer Immunotherapies. Pharmaceuticals (Basel) 2021; 14:ph14040337. [PMID: 33917617 PMCID: PMC8067507 DOI: 10.3390/ph14040337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Twenty-one styryl and phenethyl aryl ureas have been synthetized and biologically evaluated as multitarget inhibitors of Vascular endothelial growth factor receptor-2 VEGFR-2 and programmed death-ligand-1 (PD-L1) proteins in order to overcome resistance phenomena offered by cancer. The antiproliferative activity of these molecules on several tumor cell lines (HT-29, MCF-7, HeLa and A549), on the endothelial cell line human microvascular endothelial cells (HMEC)-1 and on the non-tumor cell line human embryonic kidney cells (HEK)-293 has been determined. Some derivatives were evaluated for their antiangiogenic properties such as their ability to inhibit microvessel formation using HMEC-1 or their effect on VEGFR-2 in both cancer and endothelial cell lines. In addition, the immunomodulator action of a number of selected compounds was also studied on PD-L1 and c-Myc proteins. Compounds 16 and 23 (Z) and (E)-styryl p-bromophenyl urea, respectively, showed better results than sorafenib in down-regulation of VEGFR-2 and also improved the effect of the anti-PD-L1 compound BMS-8 on both targets, PD-L1 and c-Myc proteins.
Collapse
|
207
|
Bao X, Shen N, Lou Y, Yu H, Wang Y, Liu L, Tang Z, Chen X. Enhanced anti-PD-1 therapy in hepatocellular carcinoma by tumor vascular disruption and normalization dependent on combretastatin A4 nanoparticles and DC101. Theranostics 2021; 11:5955-5969. [PMID: 33897892 PMCID: PMC8058708 DOI: 10.7150/thno.58164] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Anti-programmed cell death protein 1 (PD-1) therapy has shown promising efficacy in hepatocellular carcinoma (HCC), but its response rates in advanced HCC are lower than 20%. A critical reason for this is the imbalance between CD8+ T cells and tumor burden. Here, a novel concept of vascular disruption and normalization dependent on a polymeric vascular disrupting agent (VDA) poly (L-glutamic acid)-graft-methoxy poly (ethylene glycol)/combretastatin A4 (CA4-NPs) + a vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) inhibitor DC101 is applied to improve anti-PD-1 therapy, wherein CA4-NPs reduce tumor burden and DC101 simultaneously increases the number of intratumoral CD8+ T cells, successfully regulating the abovementioned imbalance in an H22 tumor model. Methods: Blood vessel density, tumor cell proliferation, and necrosis were evaluated to reveal the effects on reducing tumor burden by CA4-NP treatment. Pericyte coverage of blood vessels, tumor blood vessel perfusion, tumor hypoxia, and intratumoral immune cells were examined to verify their role in vascular normalization and immune cell homing of DC101. Furthermore, the effects of CA4-NPs + DC101 on reducing tumor burden and increasing the number of immune cells were studied. Finally, tumor suppression, intratumoral CD8+ T cell activation, and the synergistic effects of anti-PD-1 combined with CA4-NPs + DC101 were verified. Results: The tumor inhibition rate of anti-PD-1 antibody combined with CA4-NPs + DC101 reached 86.4%, which was significantly higher than that of anti-PD-1 (16.8%) alone. Importantly, the Q value reflecting the synergy between CA4-NPs + DC101 and anti-PD-1 was 1.24, demonstrating a strong synergistic effect. Furthermore, CA4-NPs + DC101 improved anti-PD-1 therapy by increasing the number of intratumoral CD8+ T cells (anti-PD-1, 0.31% vs triple drug combination, 1.18%). Conclusion: These results reveal a novel approach to enhance anti-PD-1 therapy with VDAs + VEGF/VEGFR2 inhibitors in HCC.
Collapse
Affiliation(s)
- Xin Bao
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Yan Lou
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Yue Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Linlin Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| |
Collapse
|
208
|
Ollauri-Ibáñez C, Ayuso-Íñigo B, Pericacho M. Hot and Cold Tumors: Is Endoglin (CD105) a Potential Target for Vessel Normalization? Cancers (Basel) 2021; 13:1552. [PMID: 33800564 PMCID: PMC8038031 DOI: 10.3390/cancers13071552] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Tumors are complex masses formed by malignant but also by normal cells. The interaction between these cells via cytokines, chemokines, growth factors, and enzymes that remodel the extracellular matrix (ECM) constitutes the tumor microenvironment (TME). This TME can be determinant in the prognosis and the response to some treatments such as immunotherapy. Depending on their TME, two types of tumors can be defined: hot tumors, characterized by an immunosupportive TME and a good response to immunotherapy; and cold tumors, which respond poorly to this therapy and are characterized by an immunosuppressive TME. A therapeutic strategy that has been shown to be useful for the conversion of cold tumors into hot tumors is vascular normalization. In this review we propose that endoglin (CD105) may be a useful target of this strategy since it is involved in the three main processes involved in the generation of the TME: angiogenesis, inflammation, and cancer-associated fibroblast (CAF) accumulation. Moreover, the analysis of endoglin expression in tumors, which is already used in the clinic to study the microvascular density and that is associated with worse prognosis, could be used to predict a patient's response to immunotherapy.
Collapse
Affiliation(s)
| | | | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Group of Physiopathology of the Vascular Endothelium (ENDOVAS), Biomedical Research Institute of Salamanca (IBSAL), Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain; (C.O.-I.); (B.A.-Í.)
| |
Collapse
|
209
|
Guo J, Zeng H, Liu Y, Shi X, Liu Y, Liu C, Chen Y. Multicomponent thermosensitive lipid complexes enhance desmoplastic tumor therapy through boosting anti-angiogenesis and synergistic strategy. Int J Pharm 2021; 601:120533. [PMID: 33781886 DOI: 10.1016/j.ijpharm.2021.120533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 01/05/2023]
Abstract
Currently, the chemotherapy drugs-loaded thermosensitive liposomes have not shown an over standard of clinical effects compared to preclinical trials. In addition to the limiting factors of clinical trial design and heating device, abnormal angiogenesis in desmoplastic tumor is a key factor for unexpected clinical efficacy. Malformed tumor vasculature may result in reduced vascular transport and the heterogeneous distribution of thermosensitive liposomes in tumor. Here, we report an anti-angiogenesis strategy through hypoxia-inducible factors (HIF)-1α-vascular endothelial growth factor (VEGF) axis based on icaritin and coix seed oil dual loaded multicomponent thermosensitive lipid complexes (IC-ML). IC-ML could downregulate the HIF-1α expression in HepG2 cells with a synergetic antitumor effect. In addition, HepG2 + LX-2 cells co-cultured 3D tumor spheres administered IC-ML showed the strongest penetration and inhibition of growth. Accordingly, IC-ML displayed improved tumor penetration and superior synergistic antitumor efficacy with HIF-1α-VEGF downregulation in vivo under mild hyperthermia. The improvement of antitumor efficacy of IC-ML comes from the anti-angiogenesis strategy and comprehensive tumor microenvironment remodeling, including depletion of cancer-associated fibroblasts as well as inhibition of M2-type tumor associated macrophage infiltration in desmoplastic tumor. This study proposes a novel multicomponent synergistic antitumor strategy to improve the therapeutic potential of thermosensitive lipid complexes for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jian Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Huating Zeng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yimin Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xinmeng Shi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Congyan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
210
|
王 婧, 彭 文, 江 美, 邬 麟. [Research Progress of Anti-angiogenic Agents Combined with Immunotherapy
in Patients with Advanced Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:196-203. [PMID: 33819970 PMCID: PMC8143969 DOI: 10.3779/j.issn.1009-3419.2021.101.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 12/05/2022]
Abstract
Lung cancer has the highest incidence rate and mortality in China, even in the world, and non-small cell lung cancer (NSCLC) accounts for about 85%. The growth and metastasis of tumor depend on the generation of blood vessels, and anti-angiogenic therapy is playing an increasingly important role, however, no significant improvement was observed in the underwent anti-angiogenic agents used for patients alone. In recent years, the application of immune checkpoint inhibitor (ICI) has significantly improved the prognosis of some lung cancer patients, however, the objective response rate of patients receiving ICI alone is low. While anti-angiogenic agents and ICI both regulate the tumor immune microenvironment and have a potential synergistic mechanism, showing a bright prospect in the combined application of anti-tumor therapy. In this review, we focused on the research and application of anti-angiogenic agents in combination with ICI in advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- 婧怡 王
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| | - 文颖 彭
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| | - 美林 江
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| | - 麟 邬
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| |
Collapse
|
211
|
Yan LH, Liu XL, Mo SS, Zhang D, Mo XW, Tang WZ. OX40 as a novel target for the reversal of immune escape in colorectal cancer. Am J Transl Res 2021; 13:923-934. [PMID: 33841630 PMCID: PMC8014382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 04/11/2020] [Indexed: 06/12/2023]
Abstract
First-generation immunological checkpoint inhibitors, such as CTLA-4, PD-L1 and PD-1 exhibit significant advantages over conventional cytotoxic drugs, such as oxaliplatin and 5-FU, for the treatment of colorectal cancer. However, these inhibitors are not ideal due to their low objective response rate and the vulnerability of these treatment methods when faced with emerging drug resistant cancers. This study summarizes the immunological characteristics of colorectal cancer treatment, and analyzes the ways in which OX40 may improve the efficacy of these treatments. Activation of the OX40 signaling pathway can enhance the activity of CD4+/CD8+ T cells and inhibit the function of Treg. Simultaneously, OX40 can directly inhibit the expression of Foxp3, affect the inhibitory function of Treg, and inhibit the immunosuppressive factors in the tumor microenvironment so as to reverse immune escape and reverse drug resistance. Therefore, OX40 is an important target for treating colorectal cancer in "cold tumors" with less immunogenicity.
Collapse
Affiliation(s)
- Lin-Hai Yan
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer HospitalNanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal CancerNanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiao-Liang Liu
- Department of Surgery, Guangxi Medical University Cancer HospitalNanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Si-Si Mo
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer HospitalNanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal CancerNanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Di Zhang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer HospitalNanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal CancerNanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xian-Wei Mo
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer HospitalNanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal CancerNanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Wei-Zhong Tang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer HospitalNanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal CancerNanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
212
|
Kim S, Park J, Kim J, Jeon JS. Microfluidic Tumor Vasculature Model to Recapitulate an Endothelial Immune Barrier Expressing FasL. ACS Biomater Sci Eng 2021; 7:1230-1241. [PMID: 33586426 DOI: 10.1021/acsbiomaterials.0c01542] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fas ligand (FasL, CD178) is known to bind to its receptor (Fas, CD95) and mediate cellular apoptosis to maintain immune homeostasis. Recently, it has been recognized that tumor cells and their microenvironments allow an adjacent vascular endothelium to express the FasL on its cell membrane, utilizing the endothelium as an immune barrier to kill antitumor cytotoxic T cells. Here, a microfluidic tumor vasculature model is presented, which enables the recapitulation of an endothelial immune barrier expressing FasL. The in vitro three-dimensional model replicates enhanced endothelial FasL expression under the hypoxic tumor microenvironment. Apoptosis rates of FasL-susceptible target cells are augmented under the microenvironment with upregulated FasL but are consequently abrogated by administrations of pharmacological inhibitions, FasL-Fas blockades. The microfluidic system suggests its promising applications in elucidating complex immunosuppressive mechanisms of the tumor microenvironment and screening of cell-mediated immunotherapies as a preclinical model.
Collapse
Affiliation(s)
- Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Joonha Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jeongsik Kim
- National Creative Research Initiatives Center, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.,KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
213
|
Powles T, Atkins MB, Escudier B, Motzer RJ, Rini BI, Fong L, Joseph RW, Pal SK, Sznol M, Hainsworth J, Stadler WM, Hutson TE, Ravaud A, Bracarda S, Suarez C, Choueiri TK, Reeves J, Cohn A, Ding B, Leng N, Hashimoto K, Huseni M, Schiff C, McDermott DF. Efficacy and Safety of Atezolizumab Plus Bevacizumab Following Disease Progression on Atezolizumab or Sunitinib Monotherapy in Patients with Metastatic Renal Cell Carcinoma in IMmotion150: A Randomized Phase 2 Clinical Trial. Eur Urol 2021; 79:665-673. [PMID: 33678522 DOI: 10.1016/j.eururo.2021.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 01/05/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND The use of immune checkpoint inhibitors combined with vascular endothelial growth factor (VEGF)-targeted therapy as second-line treatment for metastatic clear cell renal cancer (mRCC) has not been evaluated prospectively. OBJECTIVE To evaluate the efficacy and safety of atezolizumab + bevacizumab following disease progression on atezolizumab or sunitinib monotherapy in patients with mRCC. DESIGN, SETTING, AND PARTICIPANTS IMmotion150 was a multicenter, randomized, open-label, phase 2 study of patients with untreated mRCC. Patients randomized to the atezolizumab or sunitinib arm who had investigator-assessed progression as per RECIST 1.1 could be treated with second-line atezolizumab + bevacizumab. INTERVENTION Patients received atezolizumab 1200 mg intravenously (IV) plus bevacizumab 15 mg/kg IV every 3 wk following disease progression on either atezolizumab or sunitinib monotherapy. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The secondary endpoints analyzed during the second-line part of IMmotion150 included objective response rate (ORR), progression-free survival (PFS), and safety. PFS was examined using Kaplan-Meier methods. RESULTS AND LIMITATIONS Fifty-nine patients in the atezolizumab arm and 78 in the sunitinib arm were eligible, and 103 initiated second-line atezolizumab + bevacizumab (atezolizumab arm, n = 44; sunitinib arm, n = 59). ORR (95% confidence interval [CI]) was 27% (19-37%). The median PFS (95% CI) from the start of second line was 8.7 (5.6-13.7) mo. The median event follow-up duration was 19.4 (12.9-21.9) mo among the 25 patients without a PFS event. Eighty-six (83%) patients had treatment-related adverse events; 31 of 103 (30%) had grade 3/4 events. Limitations were the small sample size and selection for progressors. CONCLUSIONS The atezolizumab + bevacizumab combination had activity and was tolerable in patients with progression on atezolizumab or sunitinib. Further studies are needed to investigate sequencing strategies in mRCC. PATIENT SUMMARY Patients with advanced kidney cancer whose disease had worsened during treatment with atezolizumab or sunitinib began second-line treatment with atezolizumab + bevacizumab. Tumors shrank in more than one-quarter of patients treated with this combination, and side effects were manageable.
Collapse
Affiliation(s)
- Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | | | | | - Brian I Rini
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lawrence Fong
- University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | | | - Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | | | | | - Thomas E Hutson
- Texas Oncology-Baylor Charles A. Sammons Cancer Center, Dallas, TX, USA
| | - Alain Ravaud
- CHU Hopitaux de Bordeaux, Hôpital Saint-André, Bordeaux, France
| | | | - Cristina Suarez
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - James Reeves
- Florida Cancer Specialists & Research Institute, Fort Myers, FL, USA
| | - Allen Cohn
- Rocky Mountain Cancer Center, Denver, CO, USA
| | | | - Ning Leng
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
214
|
Takakura N. Vascular reconstitution in the tumor for more effective tumor immunotherapy. Cancer Sci 2021; 112:1348-1356. [PMID: 33587826 PMCID: PMC8019202 DOI: 10.1111/cas.14854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/09/2021] [Accepted: 02/13/2021] [Indexed: 12/12/2022] Open
Abstract
It has been widely accepted that the regulation of the tumor microenvironment is an important strategy in cancer treatment. Particularly, control of the tumor vasculature has been suggested to be critical for antitumor immunotherapy. Effectiveness of cancer immunotherapy depends on the quality and quantity of immune cells infiltrating into tumor tissues, which may be affected by the status of the tumor vasculature. Under physiological conditions, immune cells migrate from the intravascular lumen into the parenchyma especially by passing through the vascular wall of venulae. Extravasation of immune cells is induced from venulae where endothelial cells (ECs) are fully covered with pericytes from the basal side. Interaction of pericytes with ECs contributes to immune cell extravasation by several steps, ie, adhesion of immune cells to intraluminal ECs, transmigration, and chemotaxis of immune cells. Blood vessels are structurally immature and non‐functional in tumors, and therefore, induction of maturation in the tumor vasculature is a promising strategy for effective cancer therapies and is relevant not only for immune cell migration but also drug delivery.
Collapse
Affiliation(s)
- Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
215
|
Ou DL, Chen CW, Hsu CL, Chung CH, Feng ZR, Lee BS, Cheng AL, Yang MH, Hsu C. Regorafenib enhances antitumor immunity via inhibition of p38 kinase/Creb1/Klf4 axis in tumor-associated macrophages. J Immunother Cancer 2021; 9:e001657. [PMID: 33753566 PMCID: PMC7986673 DOI: 10.1136/jitc-2020-001657] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Regorafenib and other multikinase inhibitors may enhance antitumor efficacy of anti-program cell death-1 (anti-PD1) therapy in hepatocellular carcinoma (HCC). Its immunomodulatory effects, besides anti-angiogenesis, were not clearly defined. METHODS In vivo antitumor efficacy was tested in multiple syngeneic liver cancer models. Murine bone marrow-derived macrophages (BMDMs) were tested in vitro for modulation of polarization by regorafenib and activation of cocultured T cells. Markers of M1/M2 polarization were measured by quantitative reverse transcription PCR (RT-PCR), arginase activity, flow cytometry, and ELISA. Knockdown of p38 kinase and downstream Creb1/Klf4 signaling on macrophage polarization were confirmed by using knockdown of the upstream MAPK14 kinase, chemical p38 kinase inhibitor, and chromatin immunoprecipitation. RESULTS Regorafenib (5 mg/kg/day, corresponding to about half of human clinical dosage) inhibited tumor growth and angiogenesis in vivo similarly to DC-101 (anti-VEGFR2 antibody) but produced higher T cell activation and M1 macrophage polarization, increased the ratio of M1/M2 polarized BMDMs and proliferation/activation of cocultured T cells in vitro, indicating angiogenesis-independent immunomodulatory effects. Suppression of p38 kinase phosphorylation and downstream Creb1/Klf4 activity in BMDMs by regorafenib reversed M2 polarization. Regorafenib enhanced antitumor efficacy of adoptively transferred antigen-specific T cells. Synergistic antitumor efficacy between regorafenib and anti-PD1 was associated with multiple immune-related pathways in the tumor microenvironment. CONCLUSION Regorafenib may enhance antitumor immunity through modulation of macrophage polarization, independent of its anti-angiogenic effects. Optimization of regorafenib dosage for rational design of combination therapy regimen may improve the therapeutic index in the clinic.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antineoplastic Agents/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/immunology
- Cell Line, Tumor
- Coculture Techniques
- Cyclic AMP Response Element-Binding Protein/metabolism
- Kruppel-Like Factor 4/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/enzymology
- Liver Neoplasms/immunology
- Lymphocyte Activation/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/enzymology
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Phenotype
- Phenylurea Compounds/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyridines/pharmacology
- Signal Transduction
- Tumor Microenvironment
- Tumor-Associated Macrophages/drug effects
- Tumor-Associated Macrophages/enzymology
- Tumor-Associated Macrophages/immunology
- p38 Mitogen-Activated Protein Kinases/metabolism
- Mice
Collapse
Affiliation(s)
- Da-Liang Ou
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Wei Chen
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Lang Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Hung Chung
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Zi-Rui Feng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Bin-Shyun Lee
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chiun Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
216
|
Huang D, Sun L, Huang L, Chen Y. Nanodrug Delivery Systems Modulate Tumor Vessels to Increase the Enhanced Permeability and Retention Effect. J Pers Med 2021; 11:124. [PMID: 33672813 PMCID: PMC7917988 DOI: 10.3390/jpm11020124] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022] Open
Abstract
The use of nanomedicine for antitumor therapy has been extensively investigated for a long time. Enhanced permeability and retention (EPR) effect-mediated drug delivery is currently regarded as an effective way to bring drugs to tumors, especially macromolecular drugs and drug-loaded pharmaceutical nanocarriers. However, a disordered vessel network, and occluded or embolized tumor blood vessels seriously limit the EPR effect. To augment the EPR effect and improve curative effects, in this review, we focused on the perspective of tumor blood vessels, and analyzed the relationship among abnormal angiogenesis, abnormal vascular structure, irregular blood flow, extensive permeability of tumor vessels, and the EPR effect. In this commentary, nanoparticles including liposomes, micelles, and polymers extravasate through the tumor vasculature, which are based on modulating tumor vessels, to increase the EPR effect, thereby increasing their therapeutic effect.
Collapse
Affiliation(s)
- Dong Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lingna Sun
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Yanzuo Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
217
|
Cui Q, Hu Y, Ma D, Liu H. A Retrospective Observational Study of Anlotinib in Patients with Platinum-Resistant or Platinum-Refractory Epithelial Ovarian Cancer. Drug Des Devel Ther 2021; 15:339-347. [PMID: 33536747 PMCID: PMC7850384 DOI: 10.2147/dddt.s286529] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022] Open
Abstract
Objective Anlotinib, an oral small-molecular tyrosine kinase inhibitor (TKI) on tumor angiogenesis and growth, has a wide spectrum of inhibitory effects on targets such as vascular endothelial growth factor receptors 2/3 (VEGFR2/3), etc. The efficacy and safety of anlotinib in the treatment of platinum-resistant or platinum-refractory ovarian cancer were evaluated. Patients and Methods Patients with platinum-resistant or platinum-refractory ovarian cancer that treated with anlotinib in the Affiliated Cancer Hospital of Zhengzhou University from May 2018 to March 2020 were included. Medical records were reviewed in terms of objective response, survival outcomes, and safety. Results A total of 38 patients were analyzed. The median progression-free survival and the median overall survival were 7.7 months (95% CI: 6.7–8.7) and 16.5 months (95% CI: 13.3–19.7), respectively. About 17 patients received anlotinib monotherapy, and the median progression-free survival was 7.7 months (95% CI: 6.3–9.1). A total of 19 cases received anlotinib plus chemotherapy with a median progression-free survival of 8.0 months (95% CI: 4.8–11.2). A total of 2 cases received anlotinib plus anti-PD-1 antibody pembrolizumab, and 1 case had partial response, the other progressive disease. The objective response rate was 42.1% while the disease control rate was 86.8%. A total of 5 patients experienced dose reduction from 12 mg to 10 mg because of adverse effects. The most common adverse effects were hypertension (31.6%), fatigue (28.9%), anorexia (26.3%) and hand-foot syndrome (23.7%). No treatment-related deaths were recorded. Conclusion Anlotinib produced moderate improvements in progression-free survival and overall survival in patients with platinum-resistant or platinum-refractory ovarian cancer. It indicates that anlotinib maybe a new treatment option for patients with platinum-resistant or platinum-refractory ovarian cancer.
Collapse
Affiliation(s)
- Qingli Cui
- Department of Integrated Traditional and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou City, Henan Province, People's Republic of China
| | - Yanhui Hu
- Department of Integrated Traditional and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou City, Henan Province, People's Republic of China
| | - Dongyang Ma
- Department of Integrated Traditional and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou City, Henan Province, People's Republic of China
| | - Huaimin Liu
- Department of Integrated Traditional and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou City, Henan Province, People's Republic of China
| |
Collapse
|
218
|
Mpekris F, Panagi M, Voutouri C, Martin JD, Samuel R, Takahashi S, Gotohda N, Suzuki T, Papageorgis P, Demetriou P, Pierides C, Koumas L, Costeas P, Kojima M, Ishii G, Constantinidou A, Kataoka K, Cabral H, Stylianopoulos T. Normalizing the Microenvironment Overcomes Vessel Compression and Resistance to Nano-immunotherapy in Breast Cancer Lung Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001917. [PMID: 33552852 PMCID: PMC7856901 DOI: 10.1002/advs.202001917] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/13/2020] [Indexed: 05/02/2023]
Abstract
Nano-immunotherapy regimens have high potential to improve patient outcomes, as already demonstrated in advanced triple negative breast cancer with nanoparticle albumin-bound paclitaxel and the immune checkpoint blocker (ICB) atezolizumab. This regimen, however, does not lead to cures with median survival lasting less than two years. Thus, understanding the mechanisms of resistance to and development of strategies to enhance nano-immunotherapy in breast cancer are urgently needed. Here, in human tissue it is shown that blood vessels in breast cancer lung metastases are compressed leading to hypoxia. This pathophysiology exists in murine spontaneous models of triple negative breast cancer lung metastases, along with low levels of perfusion. Because this pathophysiology is consistent with elevated levels of solid stress, the mechanotherapeutic tranilast, which decompressed lung metastasis vessels, is administered to mice bearing metastases, thereby restoring perfusion and alleviating hypoxia. As a result, the nanomedicine Doxil causes cytotoxic effects into metastases more efficiently, stimulating anti-tumor immunity. Indeed, when combining tranilast with Doxil and ICBs, synergistic effects on efficacy, with all mice cured in one of the two ICB-insensitive tumor models investigated is resulted. These results suggest that strategies to treat breast cancer with nano-immunotherapy should also include a mechanotherapeutic to decompress vessels.
Collapse
Affiliation(s)
- Fotios Mpekris
- Cancer Biophysics LaboratoryDepartment of Mechanical and Manufacturing EngineeringUniversity of CyprusNicosia1678Cyprus
| | - Myrofora Panagi
- Cancer Biophysics LaboratoryDepartment of Mechanical and Manufacturing EngineeringUniversity of CyprusNicosia1678Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics LaboratoryDepartment of Mechanical and Manufacturing EngineeringUniversity of CyprusNicosia1678Cyprus
| | - John D. Martin
- Department of BioengineeringGraduate School of EngineeringThe University of TokyoBunkyoTokyo113‐8656Japan
| | - Rekha Samuel
- Centre for Stem Cell Research (A unit of inStem Bengaluru)Christian Medical College Campus BagayamVellore560065India
| | - Shinichiro Takahashi
- Department of Hepatobiliary‐Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaChiba277‐8577Japan
| | - Naoto Gotohda
- Department of Hepatobiliary‐Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaChiba277‐8577Japan
| | - Toshiyuki Suzuki
- Department of Hepatobiliary‐Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaChiba277‐8577Japan
| | - Panagiotis Papageorgis
- Department of Life SciencesProgram in Biological SciencesEuropean University CyprusNicosia2404Cyprus
| | - Philippos Demetriou
- The Center for the Study of Haematological and other MalignanciesNicosia2032Cyprus
| | - Chryso Pierides
- The Center for the Study of Haematological and other MalignanciesNicosia2032Cyprus
| | - Laura Koumas
- The Center for the Study of Haematological and other MalignanciesNicosia2032Cyprus
- Karaiskakio FoundationNicosia2032Cyprus
| | - Paul Costeas
- The Center for the Study of Haematological and other MalignanciesNicosia2032Cyprus
- Cyprus Cancer Research InstituteNicosia2032Cyprus
| | - Motohiro Kojima
- Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaChiba277‐8577Japan
| | - Genichiro Ishii
- Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaChiba277‐8577Japan
| | - Anastasia Constantinidou
- Cyprus Cancer Research InstituteNicosia2032Cyprus
- Medical SchoolUniversity of CyprusNicosia1678Cyprus
- Bank of Cyprus Oncology CentreNicosia2012Cyprus
| | - Kazunori Kataoka
- Innovation Center of NanoMedicineKawasaki Institute of Industrial PromotionKawasakiKanagawa210‐0821Japan
- Institute for Future InitiativesThe University of TokyoBunkyoTokyo113‐0033Japan
| | - Horacio Cabral
- Department of BioengineeringGraduate School of EngineeringThe University of TokyoBunkyoTokyo113‐8656Japan
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics LaboratoryDepartment of Mechanical and Manufacturing EngineeringUniversity of CyprusNicosia1678Cyprus
| |
Collapse
|
219
|
Zhou Y, Ren X, Hou Z, Wang N, Jiang Y, Luan Y. Engineering a photosensitizer nanoplatform for amplified photodynamic immunotherapy via tumor microenvironment modulation. NANOSCALE HORIZONS 2021; 6:120-131. [PMID: 33206735 DOI: 10.1039/d0nh00480d] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Photosensitizer-based photodynamic therapy (PDT) can not only kill tumor cells by the generated cytotoxic reactive oxygen species (ROS), but also trigger immunogenic cell death (ICD) and activate an immune response for immunotherapy. However, such photodynamic immunotherapy suffers from major obstacles in the tumor microenvironment. The hypoxic microenvironment greatly weakens PDT, while the immunosuppressive tumor microenvironment caused by aberrant tumor blood vessels and indoleamine 2,3-dioxygenase (IDO) leads to a significant reduction in immunotherapy. To overcome these obstacles, herein, an engineered photosensitizer nanoplatform is designed for amplified photodynamic immunotherapy by integrating chlorin e6 (Ce6, a photosensitizer), axitinib (AXT, a tyrosine kinase inhibitor) and dextro-1-methyl tryptophan (1MT, an IDO inhibitor). In our nanoplatform, AXT improves the tumor microenvironment by normalizing tumor blood vessels, which not only promotes PDT by reducing the level of hypoxia of the tumor microenvironment, but also promotes immunotherapy through facilitating infiltration of immune effector cells into the tumor and reversing the immunosuppressive effect of vascular endothelial growth factor (VEGF). Moreover, 1MT effectively inhibits the activity of IDO, further reducing the immunosuppressive nature of the tumor microenvironment. Therefore, this nanoplatform demonstrates an amplified photodynamic immunotherapy via tumor microenvironment modulation, exhibiting outstanding therapeutic efficacy against tumor growth and metastasis with negligible side toxicity. The current concept of engineering photosensitizer nanoplatforms for overcoming photodynamic immunotherapy obstacles provides a promising strategy against tumors.
Collapse
Affiliation(s)
- Yaxin Zhou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | | | | | | | | | | |
Collapse
|
220
|
Jiang H, Tai Z, Chen Z, Zhu Q, Bao L. Clinical applicability of renin-angiotensin system inhibitors in cancer treatment. Am J Cancer Res 2021; 11:318-336. [PMID: 33575074 PMCID: PMC7868760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023] Open
Abstract
The renin-angiotensin system (RAS) regulates physiological functions of the cardiovascular system, kidneys, and other tissues. Various in vivo and in vitro studies have shown that RAS plays a pivotal role in the development of malignant tumors, while several retrospective studies have confirmed that patients undergoing long-term RAS inhibitors (RASi) treatment have a lowered risk of cancer. Moreover, blocking RAS has been shown to inhibit tumor growth, metastasis, and angiogenesis in various experimental models of malignant tumors. Herein, we review the available RASi-related literature and provide an analysis using the scientific atlas software VOSviewer. We observed that recent studies have primarily focused on gene expression, tumor biology, and survival analysis. Through an in-depth data analysis from the Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx), we identified the impact of AGTR1, an essential component of RAS, on tumors, and we discuss the underlying biological mechanism of RASi. Furthermore, we outline the research progress and potential use of RASi in tumor treatment. Overall, RASi may be a promising adjunct in cancer therapy.
Collapse
Affiliation(s)
- Huirong Jiang
- Department of Pharmacy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical UniversityShanghai 200438, China
- Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
- Department of Pharmacy, Changhai Hospital, Second Military Medical UniversityShanghai 200433, China
- Bengbu Medical CollegeBengbu 233030, China
| | - Zongguang Tai
- Department of Pharmacy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical UniversityShanghai 200438, China
- Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
- Department of Pharmacy, Changhai Hospital, Second Military Medical UniversityShanghai 200433, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Leilei Bao
- Department of Pharmacy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical UniversityShanghai 200438, China
| |
Collapse
|
221
|
Wang Y, Dong J, Quan Q, Liu S, Chen X, Cai X, Qiu H, Zhang B, Guo G. Immune Cell Infiltration of the Primary Tumor Microenvironment Predicted the Treatment Outcome of Chemotherapy With or Without Bevacizumab in Metastatic Colorectal Cancer Patients. Front Oncol 2021; 10:581051. [PMID: 33585196 PMCID: PMC7873592 DOI: 10.3389/fonc.2020.581051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/08/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND With the interest in cancer immunotherapy, it may be possible to combine immunotherapy with bevacizumab and chemotherapy. We evaluated whether tumor-infiltrating immune cells are associated with the efficacy of chemotherapy with or without bevacizumab for the treatment of metastatic colorectal cancer (mCRC). METHODS This study enrolled mCRC patients on standard treatment with available detailed data and tumor tissue at Sun Yat-sen University Cancer Center between July 1, 2005, and October 1, 2017. CD3+ and CD8+ T cell densities examined by immunohistochemistry in both the tumor core (CT) and invasive margin (IM) were summed as the Immunoscore, and the CD8+/CD3+ T cell ratio was calculated. The predictive and prognostic efficacies of tumor-infiltrating immune cells for progression-free survival (PFS) and overall survival (OS) were analyzed with Kaplan-Meier and Cox analyses. RESULTS The CD8+/CD3+ T cell ratio in the microenvironment was an independent prognostic factor for OS (28.12 mo vs. 16.56 mo, P = 0.017) among the 108 studied patients. In the chemotherapy only group, patients with a high Immunoscore had a high overall response rate (ORR, 40.0% vs. 60.0%, P = 0.022), those with a low CD8+/CD3+ T cell ratio in the microenvironment had a significantly longer PFS (8.64 mo vs. 6.01 mo, P = 0.017), and those with a high CD3+ T cell density in the CT had a longer OS (16.56 mo vs. 25.66 mo, P = 0.029). In the chemotherapy combined with bevacizumab group, patients with a higher CD8+ T cell density in the IM had a longer PFS (7.62 mo vs. 11.66 mo, P = 0.034) and OS (14.55 mo vs. 23.72 mo, P = 0.033). CONCLUSION Immune cells in primary tumors play an important role in predicting mCRC treatment efficacy. CD8 predicts the effect of bevacizumab plus chemotherapy, while CD3 and CD8/CD3 predict chemotherapy efficacy.
Collapse
Affiliation(s)
- Yixing Wang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Dong
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qi Quan
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shousheng Liu
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuxing Chen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuyu Cai
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huijuan Qiu
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bei Zhang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guifang Guo
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
222
|
Park S, Oh JH, Park DJ, Zhang H, Noh M, Kim Y, Kim YS, Kim H, Kim YM, Ha SJ, Kwon YG. CU06-1004-Induced Vascular Normalization Improves Immunotherapy by Modulating Tumor Microenvironment via Cytotoxic T Cells. Front Immunol 2021; 11:620166. [PMID: 33584714 PMCID: PMC7874050 DOI: 10.3389/fimmu.2020.620166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/10/2020] [Indexed: 01/16/2023] Open
Abstract
Blocking the immune evasion mechanism of tumor cells has become an attractive means for treating cancers. However, the usage of a drug such as nivolumab (αPD-1), which blocks programmed cell death protein 1 (PD-1), turned out to be only effective against certain types of cancer. Especially, vascular abnormal structures of which deter delivery route by leakage and cause the poor perfusion were considered to be environment unfavorable to T cells and immune checkpoint blockade (ICB) delivery within the tumor microenvironment (TME). Herein, we report stabilization of tumor blood vessels by endothelial dysfunctional blocker CU06-1004, which modified the TME and showed synergistic effects with immunotherapy anti-PD-1 antibody. CU06-1004 combination therapy consistently prolonged the survival of tumor-bearing mice by decreasing tumor growth. T-cell infiltration increased in the tumors of the combination group, with cytotoxic CD8+ T cell activity within the tumor parenchyma upregulated compared with anti-PD-1 monotherapy. Tumor inhibition was associated with reduced hypoxia and reduced vessel density in the central region of the tumor. These effects correlated significantly with enhanced expression of IFN gamma and PD-L1 in tumors. Taken together, our findings suggest that CU06-1004 is a potential candidate drug capable of improving therapeutic efficacy of anti-PD-1 through beneficial changes in the TME.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/pathology
- Animals
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Apoptosis/drug effects
- Capillary Permeability/drug effects
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/drug therapy
- Cell Hypoxia/drug effects
- Cell Line, Tumor
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/pathology
- Drug Screening Assays, Antitumor
- Drug Synergism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Immune Checkpoint Inhibitors/administration & dosage
- Immune Checkpoint Inhibitors/pharmacokinetics
- Immune Checkpoint Inhibitors/therapeutic use
- Immunotherapy/methods
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/physiopathology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Saponins/administration & dosage
- Saponins/pharmacology
- Saponins/therapeutic use
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Burden/drug effects
- Tumor Escape/drug effects
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
- Songyi Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Ji Hoon Oh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Dong Jin Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Haiying Zhang
- R&D Department, Curacle Co. Ltd, Seongnam-si, South Korea
| | - Minyoung Noh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Yeomyung Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Ye-Seul Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Hyejeong Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Young-Myeong Kim
- Vascular System Research Center, Kangwon National University, Chuncheon, South Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| |
Collapse
|
223
|
Ling X, Jiang X, Li Y, Han W, Rodriguez M, Xu Z, Lin W. Sequential Treatment of Bioresponsive Nanoparticles Elicits Antiangiogenesis and Apoptosis and Synergizes with a CD40 Agonist for Antitumor Immunity. ACS NANO 2021; 15:765-780. [PMID: 33347262 PMCID: PMC8216770 DOI: 10.1021/acsnano.0c07132] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The combination of antiangiogenesis and chemotherapy regimens with cancer immunotherapy has the potential to synergistically boost antitumor immunity. Herein, we report the construction of two bioresponsive nanoparticles, namely, Podo-NP and CbP-NP, comprising prodrugs of podophyllotoxin (Podo) and carboplatin, respectively. Sequential treatment with esterase-responsive Podo-NP, redox-sensitive CbP-NP, and a CD40 agonist promotes antitumor T cell response. Podo-NP suppresses angiogenesis by preventing proliferation and migration of endothelial cells, sprouting of neovessels, formation of tubules, and stabilization of newly formed vessels. Vascular endothelial growth factor blockade and endostatin stimulation normalize tortuous tumor vasculatures to allow efficient infiltration of effector immune cells. Subsequent treatment with CbP-NP arrests the cell-division cycle and elicits the apoptosis of tumor cells. CD40 agonist activates antigen-presenting cells to process the released tumor-associated antigens from dying tumor cells, thus reversing immunosuppressive tumor microenvironments. Sequential delivery of antiangiogenic and chemotherapeutic agents with bioresponsive NPs activates tumor microenvironments and synergizes with CD40 agonist to regress transplanted tumors and inhibit disseminated tumors in a lung cancer mouse model.
Collapse
Affiliation(s)
- Xiang Ling
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Xiaomin Jiang
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Youyou Li
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Wenbo Han
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Megan Rodriguez
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Ziwan Xu
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
224
|
Zhang C, Yan Q, Li J, Zhu Y, Zhang Y. Nanoenabled Tumor Oxygenation Strategies for Overcoming Hypoxia-Associated Immunosuppression. ACS APPLIED BIO MATERIALS 2021; 4:277-294. [PMID: 35014284 DOI: 10.1021/acsabm.0c01328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy, which initiates or strengthens innate immune responses to attack cancer cells, has shown great promise in cancer treatment. However, low immune response impacted by immunosuppressive tumor microenvironment (TME) remains a key challenge, which has been found related to tumor hypoxia. Recently, nanomaterial systems are proving to be excellent platforms for tumor oxygenation, which can reverse hypoxia-associated immunosuppression, strengthen the systemic antitumor immune responses, and thus afford a striking abscopal effect to clear metastatic cancer cells. In this review, we would like to survey recent progress in utilizing nanomaterials for tumor oxygenation through approaches such as in situ O2 generation, O2 delivery, tumor vasculature normalization, and mitochondrial-respiration inhibition. Their effects on tumor hypoxia-associated immunosuppression are highlighted. We also discuss the ongoing challenges and how to further improve the clinical prospect of cancer immunotherapy.
Collapse
Affiliation(s)
- Chao Zhang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qinglong Yan
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Jiang Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Ying Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Yu Zhang
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| |
Collapse
|
225
|
Vedenko A, Panara K, Goldstein G, Ramasamy R, Arora H. Tumor Microenvironment and Nitric Oxide: Concepts and Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1277:143-158. [PMID: 33119871 DOI: 10.1007/978-3-030-50224-9_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cancer tissue exists not as a single entity, but as a combination of different cellular phenotypes which, taken together, dramatically contribute to the entirety of their ecosystem, collectively termed as the tumor microenvironment (TME). The TME is composed of both immune and nonimmune cell types, stromal components, and vasculature-all of which cooperate to promote cancer progression. Not all immune cells, however, are immune-suppressive; some of them can promote the immune microenvironment to fight the invading and uncontrollably dividing cell populations at the initial stages of tumor growth. Yet, many of these processes and cellular phenotypes fall short, and the immune ecosystem more often than not ends up stabilizing in favor of the "resistant" resident cells that begin clonal expansion and may progress to metastatic forms. Stromal components, making up the extracellular matrix and basement membrane, are also not the most innocuous: CAFs embedded throughout secrete proteases that allow the onset of one of the most invasive processes-angiogenesis-through destruction of the ECM and the basement membrane. Vasculature formation, because of angiogenesis, is the largest invader of the TME and the reason metastasis happens. Vasculature is so sporadic and omnipresent in the TME that most drug therapies are mainly focused on stopping this uncontrollable process. As the tumor continues to grow, different processes are constantly supplying it with the ingredients favorable for tumor progression and eventual metastasis. For example, angiogenesis promotes blood vessel formation that will allow the bona fide escape of tumor cells to take place. Another process like hypoxia will present itself in several forms throughout the tumor (mild or acute, cycling or permanent), starting mechanisms such as epithelial to mesenchymal transitions (EMT) of resident cells and inadvertently placing the cells in such a stressful condition that production of ROS and DNA damage is unavoidable. DNA damage can induce mutagenicity while allowing resistant cells to survive. This is where drugs and treatments can subsequently suffer in effectiveness. Finally, another molecule has just surfaced as being a very important player in the TME: nitric oxide. Often overlooked and equated with ROS and initially assigned in the category of pathogenic molecules, nitric oxide can definitely do some damage by causing metabolic reprogramming and promotion of immunosuppressive phenotypes at low concentrations. However, its actions seem to be extremely dose-dependent, and this issue has become a hot target of current treatment goals. Shockingly, nitric oxide, although omnipresent in the TME, can have a positive effect on targeting the TME broadly. Thus, while the TME is a myriad of cellular phenotypes and a combination of different tumor-promoting processes, each process is interconnected into one whole: the tumor microenvironment.
Collapse
Affiliation(s)
- Anastasia Vedenko
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Kush Panara
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Gabriella Goldstein
- College of Health Professions and Sciences, University of Central Florida, Orlando, FL, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Himanshu Arora
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
226
|
Saeed A, Park R, Sun W. The integration of immune checkpoint inhibitors with VEGF targeted agents in advanced gastric and gastroesophageal adenocarcinoma: a review on the rationale and results of early phase trials. J Hematol Oncol 2021; 14:13. [PMID: 33436042 PMCID: PMC7802258 DOI: 10.1186/s13045-021-01034-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Several targeted therapies have shown efficacy in patients with advanced gastric cancer (GC) and gastroesophageal junction adenocarcinoma (GEJC), including anti-angiogenic agents and immune checkpoint inhibitors. Ramucirumab, an anti-VEGFR2 antibody, has shown efficacy in GC, but the benefits are limited, in part due to MET-mediated resistance. Other VEGF targeted agents like VEGF tyrosine kinase inhibitors (TKIs) with broad multi-kinase inhibitory spectrum like regorafenib and cabozantinib have also shown modest single agent activity in early phase trials. For immune checkpoint inhibitors, pembrolizumab (anti-PD-1) monotherapy confers survival advantage as 3rd line therapy for the PD-L1 expressing GC and GEJC population and has been approved for use in this setting. Extensive tumor microenvironment immune modulatory effects from antiangiogenic agents have been demonstrated from preclinical data which support the clinical study rationale of dual blockade of VEGF and immune checkpoint. In addition, FDA has approved combinations of anti-VEGF/VEGFR with anti-PD-1/PD-L1 agents in hepatocellular carcinoma and renal cell carcinoma. Promising clinical activity has been demonstrated in patients with refractory GC/GEJC when treated with dual blockade combination with antiangiogenic agents and immune checkpoint inhibitors like PD-1/PD-L1 inhibitors in several phase I/II trials. This review highlights the trials investigating these novel combinations as well as their preclinical rationale.
Collapse
Affiliation(s)
- Anwaar Saeed
- Division of Medical Oncology, Department of Medicine, University of Kansas Medical Center, 2330 Shawnee Mission Pkwy, Suite 210, Westwood, Kansas City, KS, 66205, USA.
| | - Robin Park
- MetroWest Medical Center/Tufts University School of Medicine, Framingham, MA, USA
| | - Weijing Sun
- Division of Medical Oncology, Department of Medicine, University of Kansas Medical Center, 2330 Shawnee Mission Pkwy, Suite 210, Westwood, Kansas City, KS, 66205, USA
| |
Collapse
|
227
|
Attia H, Smyth E. Evolving therapies in advanced oesophago-gastric cancers and the increasing role of immunotherapy. Expert Rev Anticancer Ther 2021; 21:535-546. [PMID: 33349073 DOI: 10.1080/14737140.2021.1866548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Esophagogastric cancers remain a considerable health burden and among the top causes of global cancer-related deaths. Chemotherapy remains the cornerstone of treatment for patients with advanced disease. Doublet platinum/fluoropyrimidine therapy is established as first-line treatment with the option of adding a taxane in selected patients. Irinotecan, taxanes, and ramucirumab are approved as second-line treatments. Results from the trials KEYNOTE-059, ATTRACTION-2, and TAGS have established the use of immune checkpoint inhibitors and trifluridine/tipiracil as a third-line treatment. High PD-L1 expression, microsatellite instability, tumor mutational burden, and Epstein-Barr virus status may also be used to enrich for responses to immunotherapy. AREAS COVERED In this review, we discuss the outcome of recent trials in the later lines of therapy for esophagogastric cancer and place these in the context of current treatment paradigms. We also discuss the biology of esophagogastric cancers and how this might inform the development of new treatments. Finally, we comment on promising new drugs in development. EXPERT OPINION Recent advances in the treatment of chemo-refractory esophagogastric cancer add to the improving survival of patients with this disease. Further research is needed to improve patient selection to therapies and the earlier incorporation of these agents in the treatment journey.
Collapse
Affiliation(s)
- Hossameldin Attia
- Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Elizabeth Smyth
- Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
228
|
Pinter M, Jain RK, Duda DG. The Current Landscape of Immune Checkpoint Blockade in Hepatocellular Carcinoma: A Review. JAMA Oncol 2021; 7:113-123. [PMID: 33090190 DOI: 10.1001/jamaoncol.2020.3381] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Importance For more than a decade, sorafenib has been the only systemic treatment option for patients with advanced hepatocellular carcinoma (HCC). However, rapid progress over the past few years led to approval of other angiogenesis inhibitors and several immune checkpoint blockers (ICBs) that have been added to the treatment armamentarium for advanced HCC. Moreover, the recent success of a combination of bevacizumab with atezolizumab signals an important change in the front-line treatment of HCC. Observations This review summarizes rapidly emerging clinical data on the promise and challenges of implementing ICBs in HCC and discusses the unmet need of biomarkers to predict response or resistance to therapy. Two strategies to target immunosuppression in tumors are also discussed: one proven (vascular endothelial growth factor pathway inhibition) and one currently under investigation (transforming growth factor-β pathway inhibition). The rationale and preliminary evidence on how their inhibition may reprogram the immunosuppressive milieu and enhance the efficacy of ICBs in HCC are reviewed. Conclusion and Relevance The recent successes and failures of angiogenesis inhibitors and ICBs, alone and in combination, have provided important insights into how to implement this novel systemic therapy in HCC and led to new avenues to enhance immunotherapy efficacy in this disease.
Collapse
Affiliation(s)
- Matthias Pinter
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Dan G Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
229
|
Feldmann K, Maurer C, Peschke K, Teller S, Schuck K, Steiger K, Engleitner T, Öllinger R, Nomura A, Wirges N, Papargyriou A, Jahan Sarker RS, Ranjan RA, Dantes Z, Weichert W, Rustgi AK, Schmid RM, Rad R, Schneider G, Saur D, Reichert M. Mesenchymal Plasticity Regulated by Prrx1 Drives Aggressive Pancreatic Cancer Biology. Gastroenterology 2021; 160:346-361.e24. [PMID: 33007300 DOI: 10.1053/j.gastro.2020.09.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 08/11/2020] [Accepted: 09/06/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) is characterized by a fibroblast-rich desmoplastic stroma. Cancer-associated fibroblasts (CAFs) have been shown to display a high degree of interconvertible states including quiescent, inflammatory, and myofibroblastic phenotypes; however, the mechanisms by which this plasticity is achieved are poorly understood. Here, we aim to elucidate the role of CAF plasticity and its impact on PDAC biology. METHODS To investigate the role of mesenchymal plasticity in PDAC progression, we generated a PDAC mouse model in which CAF plasticity is modulated by genetic depletion of the transcription factor Prrx1. Primary pancreatic fibroblasts from this mouse model were further characterized by functional in vitro assays. To characterize the impact of CAFs on tumor differentiation and response to chemotherapy, various coculture experiments were performed. In vivo, tumors were characterized by morphology, extracellular matrix composition, and tumor dissemination and metastasis. RESULTS Our in vivo findings showed that Prrx1-deficient CAFs remain constitutively activated. Importantly, this CAF phenotype determines tumor differentiation and disrupts systemic tumor dissemination. Mechanistically, coculture experiments of tumor organoids and CAFs showed that CAFs shape the epithelial-to-mesenchymal phenotype and confer gemcitabine resistance of PDAC cells induced by CAF-derived hepatocyte growth factor. Furthermore, gene expression analysis showed that patients with pancreatic cancer with high stromal expression of Prrx1 display the squamous, most aggressive, subtype of PDAC. CONCLUSIONS Here, we define that the Prrx1 transcription factor is critical for tuning CAF activation, allowing a dynamic switch between a dormant and an activated state. This work shows that Prrx1-mediated CAF plasticity has significant impact on PDAC biology and therapeutic resistance.
Collapse
Affiliation(s)
- Karin Feldmann
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carlo Maurer
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Katja Peschke
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Steffen Teller
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kathleen Schuck
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Thomas Engleitner
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Rupert Öllinger
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Alice Nomura
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nils Wirges
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany
| | - Aristeidis Papargyriou
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg, Germany
| | - Rim Sabrina Jahan Sarker
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany
| | - Raphela Aranie Ranjan
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Zahra Dantes
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, Columbia University, New York, New York
| | - Roland M Schmid
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Roland Rad
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Günter Schneider
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Dieter Saur
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Maximilian Reichert
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany.
| |
Collapse
|
230
|
Li HL, Li Y, Hu HT, Shao SS, Chen CS, Guo CY, Zhao Y, Yao QJ. Clinical observation of local intervention combined with camrelizumab and apatinib in the treatment of metastatic soft-tissue sarcoma. J Cancer Res Ther 2021; 17:1718-1724. [DOI: 10.4103/jcrt.jcrt_1310_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
231
|
Furukawa N, Popel AS. Peptides that immunoactivate the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2021; 1875:188486. [PMID: 33276025 PMCID: PMC8369429 DOI: 10.1016/j.bbcan.2020.188486] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/04/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy has achieved positive clinical outcomes and is revolutionizing cancer treatment. However, cancer immunotherapy has thus far failed to improve outcomes for most "cold tumors", which are characterized by low infiltration of immune cells and immunosuppressive tumor microenvironment. Enhancing the responsiveness of cold tumors to cancer immunotherapy by stimulating the components of the tumor microenvironment is a strategy pursued in the last decade. Currently, most of the agents used to modify the tumor microenvironment are small molecules or antibodies. Small molecules exhibit low affinity and specificity towards the target and antibodies have shortcomings such as poor tissue penetration and high production cost. Peptides may overcome these drawbacks and therefore are promising materials for immunomodulating agents. Here we systematically summarize the currently developed immunoactivating peptides and discuss the potential of peptide therapeutics in cancer immunology.
Collapse
Affiliation(s)
- Natsuki Furukawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
232
|
Zhou C, Wang Y, Zhao J, Chen G, Liu Z, Gu K, Huang M, He J, Chen J, Ma Z, Feng J, Shi J, Yu X, Cheng Y, Yao Y, Chen Y, Guo R, Lin X, Wang Z, Gao G, Wang Q, Li W, Yang X, Wu L, Zhang J, Ren S. Efficacy and Biomarker Analysis of Camrelizumab in Combination with Apatinib in Patients with Advanced Nonsquamous NSCLC Previously Treated with Chemotherapy. Clin Cancer Res 2020; 27:1296-1304. [PMID: 33323401 DOI: 10.1158/1078-0432.ccr-20-3136] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/16/2020] [Accepted: 12/10/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE Our preclinical work suggests that appropriate angiogenesis inhibition could potentiate PD-1/PD-L1 blockade via alleviating hypoxia, increasing infiltration of CD8+ T cells and reducing recruitment of tumor-associated macrophages. We hereby conducted a clinical trial to evaluate this combination in pretreated patients with advanced non-small cell lung cancer (NSCLC). PATIENTS AND METHODS The study included phase Ib apatinib dose-escalation and phase II expansion cohorts. Patients received apatinib at doses of 250-500 mg orally once daily, in combination with camrelizumab 200 mg intravenously every 2 weeks. RESULTS From March 2017 to October 2018, 105 chemotherapy-pretreated patients with nonsquamous NSCLC were enrolled and received apatinib 250 mg (recommended phase II dose) and camrelizumab. Among them, one (1.0%) complete response, 28 (26.7%) partial responses, and 48 (45.7%) stable diseases were observed. In the efficacy-evaluable population (n = 94), objective response rate (ORR) was 30.9% [95% confidence interval (CI), 21.7-41.2]. The median progression-free survival was 5.7 months (95% CI, 4.5-8.8) and overall survival was 15.5 months (95% CI, 10.9-24.5). Efficacy of combination therapy was evident across all PD-L1 and tumor mutation burden subgroups, and appeared to be improved in patients with STK11/KEAP1 mutation (mutant vs. wild-type, ORR: 42.9% vs. 28.1%; 1-year survival rate: 85.1% vs. 53.1%). No unexpected adverse events were observed. CONCLUSIONS Combined apatinib and camrelizumab showed encouraging antitumor activity and acceptable toxicity in chemotherapy-pretreated patients with advanced nonsquamous NSCLC. Patients with STK11/KEAP1 mutation might derive more benefits from this combination. We will validate these results in an ongoing phase III trial (NCT04203485).
Collapse
Affiliation(s)
- Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Yina Wang
- Department of Oncology, The First Affiliated Hospital Zhejiang University, Hangzhou, P.R. China
| | - Jun Zhao
- Department of Oncology, Beijing Cancer Hospital, Beijing, P.R. China
| | - Gongyan Chen
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, P.R. China
| | - Zhihua Liu
- Thoracic Tumor Radiotherapy Department, Jiangxi Cancer Hospital, Nanchang, P.R. China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
| | - Meijuan Huang
- Department of Thoracic Oncology, West China Hospital of Sichuan University, Chengdu, P.R. China
| | - Jianxing He
- Thoracic Surgery Department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, P.R. China
| | - Jianhua Chen
- Department of Medical Oncology, Hunan Cancer Hospital, Changsha, P.R. China
| | - Zhiyong Ma
- Department of Oncology, Henan Cancer Hospital, Zhengzhou, P.R. China
| | - Jifeng Feng
- Department of Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jianhua Shi
- Department of Oncology, Linyi Cancer Hospital, Linyi, P.R. China
| | - Xinmin Yu
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, P.R. China
| | - Ying Cheng
- Department of Medical Oncology, Jilin Cancer Hospital, Changchun, P.R. China
| | - Yu Yao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yuan Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Renhua Guo
- Department of Oncology, Jiangsu Province Hospital, Nanjing, P.R. China
| | - Xiaoyan Lin
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| | - Zhehai Wang
- Department of Oncology, Shandong Cancer Hospital, Jinan, P.R. China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Quanren Wang
- Department of Clinical Research and Development, Jiangsu Hengrui Medicine Co. Ltd., Shanghai, P.R. China
| | - Weixia Li
- Department of Clinical Research and Development, Jiangsu Hengrui Medicine Co. Ltd., Shanghai, P.R. China
| | - Xinfeng Yang
- Department of Clinical Research and Development, Jiangsu Hengrui Medicine Co. Ltd., Shanghai, P.R. China
| | - Lihong Wu
- Genecast Precision Medicine Technology Institute, Beijing, P.R. China
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, Department of Cancer Biology, University of Kansas Cancer Center (KUCC), University of Kansas Medical Center (KUMC), Overland Park, Kansas
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China.
| |
Collapse
|
233
|
Veerasubramanian PK, Trinh A, Akhtar N, Liu WF, Downing TL. Biophysical and epigenetic regulation of cancer stemness, invasiveness and immune action. CURRENT TISSUE MICROENVIRONMENT REPORTS 2020; 1:277-300. [PMID: 33817661 PMCID: PMC8015331 DOI: 10.1007/s43152-020-00021-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/14/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The tumor microenvironment (TME) is an amalgam of multiple dysregulated biophysical cues that can alter cellular behavior through mechanotransductive signaling and epigenetic modifications. Through this review, we seek to characterize the extent of biophysical and epigenetic regulation of cancer stemness and tumor-associated immune cells in order to identify ideal targets for cancer therapy. RECENT FINDINGS Recent studies have identified cancer stemness and immune action as significant contributors to neoplastic disease, due to their susceptibility to microenvironmental influences. Matrix stiffening, altered vasculature, and resultant hypoxia within the TME can influence cancer stem cell (CSC) and immune cell behavior, as well as alter the epigenetic landscapes involved in cancer development. SUMMARY This review highlights the importance of aberrant biophysical cues in driving cancer progression through altered behavior of CSCs and immune cells, which in turn sustains further biophysical dysregulation. We examine current and potential therapeutic approaches that break this self-sustaining cycle of disease progression by targeting the presented biophysical and epigenetic signatures of cancer. We also summarize strategies including the normalization of the TME, targeted drug delivery, and inhibition of cancer-enabling epigenetic players.
Collapse
Affiliation(s)
- Praveen Krishna Veerasubramanian
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA, USA
| | - Annie Trinh
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA, USA
- Department of Microbiology and Molecular Genetics, University of California-Irvine, Irvine, CA, USA
| | - Navied Akhtar
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA, USA
| | - Wendy F. Liu
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California-Irvine, Irvine, CA, USA
| | - Timothy L. Downing
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA, USA
- Department of Microbiology and Molecular Genetics, University of California-Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
234
|
Leong A, Kim M. The Angiopoietin-2 and TIE Pathway as a Therapeutic Target for Enhancing Antiangiogenic Therapy and Immunotherapy in Patients with Advanced Cancer. Int J Mol Sci 2020; 21:ijms21228689. [PMID: 33217955 PMCID: PMC7698611 DOI: 10.3390/ijms21228689] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/04/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Despite significant advances made in cancer treatment, the development of therapeutic resistance to anticancer drugs represents a major clinical problem that limits treatment efficacy for cancer patients. Herein, we focus on the response and resistance to current antiangiogenic drugs and immunotherapies and describe potential strategies for improved treatment outcomes. Antiangiogenic treatments that mainly target vascular endothelial growth factor (VEGF) signaling have shown efficacy in many types of cancer. However, drug resistance, characterized by disease recurrence, has limited therapeutic success and thus increased our urgency to better understand the mechanism of resistance to inhibitors of VEGF signaling. Moreover, cancer immunotherapies including immune checkpoint inhibitors (ICIs), which stimulate antitumor immunity, have also demonstrated a remarkable clinical benefit in the treatment of many aggressive malignancies. Nevertheless, the emergence of resistance to immunotherapies associated with an immunosuppressive tumor microenvironment has restricted therapeutic response, necessitating the development of better therapeutic strategies to increase treatment efficacy in patients. Angiopoietin-2 (ANG2), which binds to the receptor tyrosine kinase TIE2 in endothelial cells, is a cooperative driver of angiogenesis and vascular destabilization along with VEGF. It has been suggested in multiple preclinical studies that ANG2-mediated vascular changes contribute to the development and persistence of resistance to anti-VEGF therapy. Further, emerging evidence suggests a fundamental link between vascular abnormalities and tumor immune evasion, supporting the rationale for combination strategies of immunotherapy with antiangiogenic drugs. In this review, we discuss the recent mechanistic and clinical advances in targeting angiopoietin signaling, focusing on ANG2 inhibition, to enhance therapeutic efficacy of antiangiogenic and ICI therapies. In short, we propose that a better mechanistic understanding of ANG2-mediated vascular changes will provide insight into the significance of ANG2 in treatment response and resistance to current antiangiogenic and ICI therapies. These advances will ultimately improve therapeutic modalities for cancer treatment.
Collapse
|
235
|
Solimando AG, Summa SD, Vacca A, Ribatti D. Cancer-Associated Angiogenesis: The Endothelial Cell as a Checkpoint for Immunological Patrolling. Cancers (Basel) 2020; 12:cancers12113380. [PMID: 33203154 PMCID: PMC7696032 DOI: 10.3390/cancers12113380] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary A clinical decision and study design investigating the level and extent of angiogenesis modulation aimed at vascular normalization without rendering tissues hypoxic is key and represents an unmet medical need. Specifically, determining the active concentration and optimal times of the administration of antiangiogenetic drugs is crucial to inhibit the growth of any microscopic residual tumor after surgical resection and in the pre-malignant and smolder neoplastic state. This review uncovers the pre-clinical translational insights crucial to overcome the caveats faced so far while employing anti-angiogenesis. This literature revision also explores how abnormalities in the tumor endothelium harm the crosstalk with an effective immune cell response, envisioning a novel combination with other anti-cancer drugs and immunomodulatory agents. These insights hold vast potential to both repress tumorigenesis and unleash an effective immune response. Abstract Cancer-associated neo vessels’ formation acts as a gatekeeper that orchestrates the entrance and egress of patrolling immune cells within the tumor milieu. This is achieved, in part, via the directed chemokines’ expression and cell adhesion molecules on the endothelial cell surface that attract and retain circulating leukocytes. The crosstalk between adaptive immune cells and the cancer endothelium is thus essential for tumor immune surveillance and the success of immune-based therapies that harness immune cells to kill tumor cells. This review will focus on the biology of the endothelium and will explore the vascular-specific molecular mediators that control the recruitment, retention, and trafficking of immune cells that are essential for effective antitumor immunity. The literature revision will also explore how abnormalities in the tumor endothelium impair crosstalk with adaptive immune cells and how targeting these abnormalities can improve the success of immune-based therapies for different malignancies, with a particular focus on the paradigmatic example represented by multiple myeloma. We also generated and provide two original bio-informatic analyses, in order to sketch the physiopathology underlying the endothelial–neoplastic interactions in an easier manner, feeding into a vicious cycle propagating disease progression and highlighting novel pathways that might be exploited therapeutically.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico-IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080-5478326 (D.R.)
| | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy;
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.G.S.); (D.R.); Tel.: +39-3395626475 (A.G.S.); +39-080-5478326 (D.R.)
| |
Collapse
|
236
|
Quintela-Fandino M, Holgado E, Manso L, Morales S, Bermejo B, Colomer R, Apala JV, Blanco R, Muñoz M, Caleiras E, Iranzo V, Martinez M, Dominguez O, Hornedo J, Gonzalez-Cortijo L, Cortes J, Gasol Cudos A, Malon D, Lopez-Alonso A, Moreno-Ortíz MC, Mouron S, Mañes S. Immuno-priming durvalumab with bevacizumab in HER2-negative advanced breast cancer: a pilot clinical trial. Breast Cancer Res 2020; 22:124. [PMID: 33176887 PMCID: PMC7661209 DOI: 10.1186/s13058-020-01362-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/25/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Preclinical research suggests that the efficacy of immune checkpoint inhibitors in breast cancer can be enhanced by combining them with antiangiogenics, particularly in a sequential fashion. We sought to explore the efficacy and biomarkers of combining the anti-PD-L1 durvalumab plus the antiangiogenic bevacizumab after bevacizumab monotherapy for advanced HER2-negative breast cancer. METHODS Patients had advanced HER2-negative disease that progressed while receiving single-agent bevacizumab maintenance as a part of a previous chemotherapy plus bevacizumab regimen. Treatment consisted of bi-weekly durvalumab plus bevacizumab (10 mg/kg each i.v.). Peripheral-blood mononuclear cells (PBMCs) were obtained before the first durvalumab dose and every 4 weeks and immunophenotyped by flow-cytometry. A fresh pre-durvalumab tumor biopsy was obtained; gene-expression studies and immunohistochemical staining to assess vascular normalization and characterize the immune infiltrate were conducted. Patients were classified as "non-progressors" if they had clinical benefit (SD/PR/CR) at 4 months. The co-primary endpoints were the changes in the percentage T cell subpopulations in PBMCs in progressors versus non-progressors, and PFS/OS time. RESULTS Twenty-six patients were accrued. Median PFS and OS were 3.5 and 11 months; a trend for a longer OS was detected for the hormone-positive subset (19.8 versus 7.4 months in triple-negatives; P = 0.11). Clinical benefit rate at 2 and 4 months was 60% and 44%, respectively, without significant differences between hormone-positive and triple-negative (P = 0.73). Non-progressors' tumors displayed vascular normalization features as a result of previous bevacizumab, compared with generally abnormal patterns observed in progressors. Non-progressors also showed increased T-effector and T-memory signatures and decreased TREG signatures in gene expression studies in baseline-post-bevacizumab-tumors compared with progressors. Notably, analysis of PBMC populations before durvalumab treatment was concordant with the findings in tumor samples and showed a decreased percentage of circulating TREGs in non-progressors. CONCLUSIONS This study reporting on sequential bevacizumab+durvalumab in breast cancer showed encouraging activity in a heavily pre-treated cohort. The correlative studies agree with the preclinical rationale supporting an immunopriming effect exerted by antiangiogenic treatment, probably by reducing TREGs cells both systemically and in tumor tissue. The magnitude of this benefit should be addressed in a randomized setting. TRIAL REGISTRATION (www.clinicaltrials.gov): NCT02802098 . Registered on June 16, 2020.
Collapse
MESH Headings
- Adult
- Aged
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Bevacizumab/administration & dosage
- Bevacizumab/adverse effects
- Breast/pathology
- Breast Neoplasms/blood
- Breast Neoplasms/drug therapy
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Disease Progression
- Female
- Humans
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Middle Aged
- Pilot Projects
- Progression-Free Survival
- Proof of Concept Study
- Receptor, ErbB-2/analysis
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain.
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain.
- Medical Oncology Department, Hospital Universitario Quiron, Pozuelo de Alarcon, Spain.
| | - Esther Holgado
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Luis Manso
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Serafin Morales
- Medical Oncology Department, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Begoña Bermejo
- Medical Oncology Department, Hospital Clínico Universitario, Valencia, Spain
- INCLIVA, Valencia, Spain
- CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Ramon Colomer
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan V Apala
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Raquel Blanco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain
| | - Manuel Muñoz
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit - Biotechnology Program, CNIO - Spanish National Cancer Research Center, Madrid, Spain
| | - Vega Iranzo
- CIBERONC, Instituto Carlos III, Madrid, Spain
- Medical Oncology Department, Hospital General Universitario de Valencia, Valencia, Spain
- Medicine Department, Universitat de Valencia, Valencia, Spain
| | - Mario Martinez
- Pathology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Orlando Dominguez
- Genomics Core Unit - Biotechnology Program, CNIO - Spanish National Cancer Research Center, Madrid, Spain
| | - Javier Hornedo
- Medical Oncology Department, Hospital Universitario Quiron, Pozuelo de Alarcon, Spain
| | | | - Javier Cortes
- ION Institute of Oncology, Quironsalud Group - Madrid & Barcelona, Barcelona, Spain
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Ariadna Gasol Cudos
- Medical Oncology Department, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Diego Malon
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Antonio Lopez-Alonso
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - María C Moreno-Ortíz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain
| | - Silvana Mouron
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain.
| |
Collapse
|
237
|
Abstract
The rapid development of nanobiotechnology has enabled progress in therapeutic cancer vaccines. These vaccines stimulate the host innate immune response by tumor antigens followed by a cascading adaptive response against cancer. However, an improved antitumor immune response is still in high demand because of the unsatisfactory clinical performance of the vaccine in tumor inhibition and regression. To date, a complicated tumor immunosuppressive environment and suboptimal design are the main obstacles for therapeutic cancer vaccines. The optimization of tumor antigens, vaccine delivery pathways, and proper adjuvants for innate immune response initiation, along with reprogramming of the tumor immunosuppressive environment, is essential for therapeutic cancer vaccines in triggering an adequate antitumor immune response. In this review, we aim to review the challenges in and strategies for enhancing the efficacy of therapeutic vaccines. We start with the summary of the available tumor antigens and their properties and then the optimal strategies for vaccine delivery. Subsequently, the vaccine adjuvants focused on the intrinsic adjuvant properties of nanostructures are further discussed. Finally, we summarize the combination strategies with therapeutic cancer vaccines and discuss their positive impact in cancer immunity.
Collapse
Affiliation(s)
- Jie Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 1001190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Muhetaerjiang Mamuti
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 1001190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 1001190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
238
|
Fan Y, Zhao J, Wang Q, Huang D, Li X, Chen J, Fang Y, Duan J, Zhou C, Hu Y, Yang H, Hu Y, Zhou J, Lin X, Wang L, Wang Z, Xu Y, Zhang T, Shi W, Zou J, Wang J. Camrelizumab Plus Apatinib in Extensive-Stage SCLC (PASSION): A Multicenter, Two-Stage, Phase 2 Trial. J Thorac Oncol 2020; 16:299-309. [PMID: 33166719 DOI: 10.1016/j.jtho.2020.10.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Treatment options in the second-line extensive-stage SCLC (ED-SCLC) setting are limited. The PASSION study (ClinicalTrials.gov identifier: NCT03417895) was a phase 2 study of camrelizumab plus apatinib in ED-SCLC after platinum-based chemotherapy. METHODS In stage I of the study, patients were randomized (1:1:1) to receive camrelizumab 200 mg every 2 weeks plus apatinib 375 mg once daily (QD), 5 days on and 2 days off, or 7 days on and 7 days off (six patients each cohort). On the basis of tolerability during the first 28-day cycle and efficacy data at stage I, one cohort was chosen to expand to 45 patients at stage II. The primary end point was objective response rate (ORR). RESULTS From April 20, 2018 to March 12, 2019, a total of 59 patients were enrolled, with 47 patients in the QD cohort. In the QD cohort, confirmed ORR reached 34.0% (95% confidence interval: 20.9‒49.3), the median progression-free survival was 3.6 months, and the median overall survival was 8.4 months. Chemotherapy-sensitive and chemotherapy-resistant patients (defined as patients with disease relapse at ≥90 and <90 d after platinum-based chemotherapy, respectively) had comparable confirmed ORR (37.5% versus 32.3%), median progression-free survival (3.6 versus 2.7 mo), and median overall survival (9.6 versus 8.0 mo). Treatment-related adverse events of grade 3 or higher were reported in 43 of 59 patients (72.9%). Five patients (8.5%) discontinued because of treatment-related adverse events. CONCLUSIONS Camrelizumab plus apatinib exhibited potential antitumor activity in patients with both chemotherapy-sensitive and chemotherapy-resistant ED-SCLC who had failed platinum-based chemotherapy with an acceptable toxicity profile. This phase 2 data warrant further clinical studies of camrelizumab plus apatinib in SCLC.
Collapse
Affiliation(s)
- Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Jun Zhao
- Department of Thoracic Medical Oncology, Beijing Cancer Hospital, Beijing, People's Republic of China
| | - Qiming Wang
- Department of Internal Medicine, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Dingzhi Huang
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Xingya Li
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jianhua Chen
- Department of Medical Oncology, Hunan Cancer Hospital, Changsha, People's Republic of China
| | - Yong Fang
- Department of Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jianchun Duan
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yanping Hu
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, People's Republic of China
| | - Haihua Yang
- Department of Radiotherapy Section, Taizhou Hospital of Zhejiang Province, Taizhou, People's Republic of China
| | - Yi Hu
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jianying Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoyan Lin
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Lifeng Wang
- Department of Medical Oncology, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Zhijie Wang
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yanjun Xu
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Tao Zhang
- Hengrui Medicine, Shanghai, People's Republic of China
| | - Wei Shi
- Hengrui Medicine, Shanghai, People's Republic of China
| | - Jianjun Zou
- Hengrui Medicine, Shanghai, People's Republic of China
| | - Jie Wang
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
| |
Collapse
|
239
|
Sundaram A, Peng L, Chai L, Xie Z, Ponraj JS, Wang X, Wang G, Zhang B, Nie G, Xie N, Rajesh Kumar M, Zhang H. Advanced nanomaterials for hypoxia tumor therapy: challenges and solutions. NANOSCALE 2020; 12:21497-21518. [PMID: 33094770 DOI: 10.1039/d0nr06271e] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
In recent years, nanomaterials and nanotechnology have emerged as vital factors in the medical field with a unique contribution to cancer medicine. Given the increasing number of cancer patients, it is necessarily required to develop innovative strategies and therapeutic modalities to tackle hypoxia, which forms a hallmark and great barrier in treating solid tumors. The present review details the challenges in nanotechnology-based hypoxia, targeting the strategies and solutions for better therapeutic performances. The interaction between hypoxia and tumor is firstly introduced. Then, we review the recently developed engineered nanomaterials towards multimodal hypoxia tumor therapies, including chemotherapy, radiotherapy, and sonodynamic treatment. In the next part, we summarize the nanotechnology-based strategies for overcoming hypoxia problems. Finally, current challenges and future directions are proposed for successfully overcoming the hypoxia tumor problems.
Collapse
Affiliation(s)
- Aravindkumar Sundaram
- Department of Orthopaedic Surgery, the Sixth Affiliated Hospital of Guangzhou Medical University, 511508 Qingyuan, Guangdong, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
García-Martínez E, Redondo A, Piulats JM, Rodríguez A, Casado A. Are antiangiogenics a good 'partner' for immunotherapy in ovarian cancer? Angiogenesis 2020; 23:543-557. [PMID: 32691290 PMCID: PMC7524856 DOI: 10.1007/s10456-020-09734-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
Abstract
Ovarian cancer (OC) is associated with poor survival because there are a limited number of effective therapies. Two processes key to OC progression, angiogenesis and immune evasion, act synergistically to promote tumor progression. Tumor-associated angiogenesis promotes immune evasion, and tumor-related immune responses in the peritoneal cavity and tumor microenvironment (TME) affect neovascular formation. Therefore, suppressing the angiogenic pathways could facilitate the arrival of immune effector cells and reduce the presence of myeloid cells involved in immune suppression. To date, clinical studies have shown significant benefits with antiangiogenic therapy as first-line therapy in OC, as well as in recurrent disease, and the vascular endothelial growth factor (VEGF) inhibitor bevacizumab is now an established therapy. Clinical data with immunomodulators in OC are more limited, but suggest that they could benefit some patients with recurrent disease. The preliminary results of two phase III trials have shown that the addition of immunomodulators to chemotherapy does not improve progression-free survival. For this reason, it could be interesting to look for synergistic effects between immunomodulators and other active drugs in OC. Since bevacizumab is approved for use in OC, and is tolerable when used in combination with immunotherapy in other indications, a number of clinical studies are underway to investigate the use of bevacizumab in combination with immunotherapeutic agents in OC. This strategy seeks to normalize the TME via the anti-VEGF actions of bevacizumab, while simultaneously stimulating the immune response via the immunotherapy. Results of these studies are awaited with interest.
Collapse
Affiliation(s)
- Elena García-Martínez
- Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Avenida Marques de los Velez, 30008, Murcia, Spain.
| | - Andres Redondo
- Medical Oncology Department, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
| | - Josep Maria Piulats
- Institut Català d'OncologiaMedical Oncology Unit - IDIBELL/OncoBell - CIBERONC, Barcelona, Spain
| | | | - Antonio Casado
- Department of Medical Oncology, Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
241
|
Kato R, Jinnouchi N, Tuyukubo T, Ikarashi D, Matsuura T, Maekawa S, Kato Y, Kanehira M, Takata R, Ishida K, Obara W. TIM3 expression on tumor cells predicts response to anti-PD-1 therapy for renal cancer. Transl Oncol 2020; 14:100918. [PMID: 33129110 PMCID: PMC7586238 DOI: 10.1016/j.tranon.2020.100918] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/27/2020] [Accepted: 10/12/2020] [Indexed: 01/05/2023] Open
Abstract
Multi-IF analysis was performed to identify novel prognostic biomarker for advanced RCC patients treated with anti-PD-1 therapy. The strongest prognostic factor of response to anti-PD-1 therapy was identified as TIM3 expression on tumor cells. Patients with TIM3-positive tumor cells showed significantly longer OS and PFS than those with TIM3-negative tumor cells.
This study aimed to identify novel prognostic biomarker for advanced renal cell carcinoma (RCC) patients treated with anti-PD-1 therapy, using quantitative multi-immunofluorescence (IF) analysis of tumor immunity. Twenty-five consecutive patients who had metastatic or unresectable RCC treated with anti-PD-1 therapy were studied. The patients were divided into a responder group (n = 12) and a non-responder group (n = 13). Quantitative multi-IF staining was performed on biopsy or surgical kidney samples using a panel of antibodies. Sections were scanned using a Mantra microscope, and the images were analyzed with inForm™ software. Responders had significantly higher rate of TIM3-positive tumor (100% versus 53.9%, p < 0.01) than non-responders. Multi-IF analysis showed that TIM3 expression on tumor cells was most strongly related to response to anti-PD-1 therapy, while some of the known immune-related prognostic factors in RCC (CD45RO, FOXP3, VEGF, PD-L1, PD-L2, CD163) had no significant association. Patients with TIM3-positive tumor showed significantly longer overall survival (not reached median time versus 6.0 months, p < 0.01) and progression-free survival (18.9 versus 1.1 months, p < 0.01) than those with TIM3-negative tumor. Immunohistochemistry study using samples obtained after anti-PD-1 therapy showed infiltration of CD163 macrophages and release of HMGB1, a ligand of TIM3, in necrotic tumor area. In conclusion, our study found clinical correlation between TIM3 expression on tumor cells and response to anti-PD-1 therapy. Further studies are warranted to verify whether TIM3 expression on tumor cells before systemic therapy predicts the efficacy of anti-PD-1 therapy for RCC in the clinical setting.
Collapse
Affiliation(s)
- Renpei Kato
- Department of Urology, Iwate Medical University, Morioka-city, Japan.
| | - Noriaki Jinnouchi
- Department of Urology, Iwate Medical University, Morioka-city, Japan
| | - Takashi Tuyukubo
- Department of Urology, Iwate Medical University, Morioka-city, Japan; Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa, Iwate, Japan
| | - Daiki Ikarashi
- Department of Urology, Iwate Medical University, Morioka-city, Japan
| | - Tomohiko Matsuura
- Department of Urology, Iwate Medical University, Morioka-city, Japan
| | | | - Yoichiro Kato
- Department of Urology, Iwate Medical University, Morioka-city, Japan
| | - Mitsugu Kanehira
- Department of Urology, Iwate Medical University, Morioka-city, Japan
| | - Ryo Takata
- Department of Urology, Iwate Medical University, Morioka-city, Japan
| | - Kazuyuki Ishida
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Shiwa, Iwate, Japan; Department of Molecular Diagnostic Pathology, Dokkyo Medical University, Shimotsuga, Tochigi, Japan
| | - Wataru Obara
- Department of Urology, Iwate Medical University, Morioka-city, Japan
| |
Collapse
|
242
|
Cook M, Chauhan A. Clinical Application of Oncolytic Viruses: A Systematic Review. Int J Mol Sci 2020; 21:ijms21207505. [PMID: 33053757 PMCID: PMC7589713 DOI: 10.3390/ijms21207505] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 02/07/2023] Open
Abstract
Leveraging the immune system to thwart cancer is not a novel strategy and has been explored via cancer vaccines and use of immunomodulators like interferons. However, it was not until the introduction of immune checkpoint inhibitors that we realized the true potential of immunotherapy in combating cancer. Oncolytic viruses are one such immunotherapeutic tool that is currently being explored in cancer therapeutics. We present the most comprehensive systematic review of all oncolytic viruses in Phase 1, 2, and 3 clinical trials published to date. We performed a systematic review of all published clinical trials indexed in PubMed that utilized oncolytic viruses. Trials were reviewed for type of oncolytic virus used, method of administration, study design, disease type, primary outcome, and relevant adverse effects. A total of 120 trials were found; 86 trials were available for our review. Included were 60 phase I trials, five phase I/II combination trials, 19 phase II trials, and two phase III clinical trials. Oncolytic viruses are feverously being evaluated in oncology with over 30 different types of oncolytic viruses being explored either as a single agent or in combination with other antitumor agents. To date, only one oncolytic virus therapy has received an FDA approval but advances in bioengineering techniques and our understanding of immunomodulation to heighten oncolytic virus replication and improve tumor kill raises optimism for its future drug development.
Collapse
Affiliation(s)
- Mary Cook
- Department of Internal Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, 22 S. Greene Street, Baltimore, MD 21201, USA;
| | - Aman Chauhan
- Department of Internal Medicine-Medical Oncology, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Center, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +504-278-0134
| |
Collapse
|
243
|
Mukaida N, Zhang D, Sasaki SI. Emergence of Cancer-Associated Fibroblasts as an Indispensable Cellular Player in Bone Metastasis Process. Cancers (Basel) 2020; 12:2896. [PMID: 33050237 PMCID: PMC7600711 DOI: 10.3390/cancers12102896] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Bone metastasis is frequently complicated in patients with advanced solid cancers such as breast, prostate and lung cancers, and impairs patients' quality of life and prognosis. At the first step of bone metastasis, cancer cells adhere to the endothelium in bone marrow and survive in a dormant state by utilizing hematopoietic niches present therein. Once a dormant stage is disturbed, cancer cells grow through the interaction with various bone marrow resident cells, particularly osteoclasts and osteoblasts. Consequently, osteoclast activation is a hallmark of bone metastasis. As a consequence, the drugs targeting osteoclast activation are frequently used to treat bone metastasis but are not effective to inhibit cancer cell growth in bone marrow. Thus, additional types of resident cells are presumed to contribute to cancer cell growth in bone metastasis sites. Cancer-associated fibroblasts (CAFs) are fibroblasts that accumulate in cancer tissues and can have diverse roles in cancer progression and metastasis. Given the presence of CAFs in bone metastasis sites, CAFs are emerging as an important cellular player in bone metastasis. Hence, in this review, we will discuss the potential roles of CAFs in tumor progression, particularly bone metastasis.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan; (D.Z.); (S.S.)
| | | | | |
Collapse
|
244
|
Chang CC, Dinh TK, Lee YA, Wang FN, Sung YC, Yu PL, Chiu SC, Shih YC, Wu CY, Huang YD, Wang J, Lu TT, Wan D, Chen Y. Nanoparticle Delivery of MnO 2 and Antiangiogenic Therapy to Overcome Hypoxia-Driven Tumor Escape and Suppress Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44407-44419. [PMID: 32865389 DOI: 10.1021/acsami.0c08473] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Antiangiogenic therapy is widely administered in many cancers, and the antiangiogenic drug sorafenib offers moderate benefits in advanced hepatocellular carcinoma (HCC). However, antiangiogenic therapy can also lead to hypoxia-driven angiogenesis and immunosuppression in the tumor microenvironment (TME) and metastasis. Here, we report the synthesis and evaluation of NanoMnSor, a tumor-targeted, nanoparticle drug carrier that efficiently codelivers oxygen-generating MnO2 and sorafenib into HCC. We found that MnO2 not only alleviates hypoxia by catalyzing the decomposition of H2O2 to oxygen but also enhances pH/redox-responsive T1-weighted magnetic resonance imaging and drug-release properties upon decomposition into Mn2+ ions in the TME. Moreover, macrophages exposed to MnO2 displayed increased mRNA associated with the immunostimulatory M1 phenotype. We further show that NanoMnSor treatment leads to sorafenib-induced decrease in tumor vascularization and significantly suppresses primary tumor growth and distal metastasis, resulting in improved overall survival in a mouse orthotopic HCC model. Furthermore, NanoMnSor reprograms the immunosuppressive TME by reducing the hypoxia-induced tumor infiltration of tumor-associated macrophages, promoting macrophage polarization toward the immunostimulatory M1 phenotype, and increasing the number of CD8+ cytotoxic T cells in tumors, thereby augmenting the efficacy of anti-PD-1 antibody and whole-cell cancer vaccine immunotherapies. Our study demonstrates the potential of oxygen-generating nanoparticles to deliver antiangiogenic agents, efficiently modulate the hypoxic TME, and overcome hypoxia-driven drug resistance, thereby providing therapeutic benefit in cancer.
Collapse
Affiliation(s)
- Chih-Chun Chang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Trinh Kieu Dinh
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-An Lee
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Fu-Nien Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Pei-Lun Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Shao-Chieh Chiu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yu-Chuan Shih
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Cheng-Yun Wu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-Da Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Dehui Wan
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
245
|
Choi YS, Jang H, Gupta B, Jeong JH, Ge Y, Yong CS, Kim JO, Bae JS, Song IS, Kim IS, Lee YM. Tie2-mediated vascular remodeling by ferritin-based protein C nanoparticles confers antitumor and anti-metastatic activities. J Hematol Oncol 2020; 13:123. [PMID: 32928251 PMCID: PMC7489044 DOI: 10.1186/s13045-020-00952-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Conventional therapeutic approaches for tumor angiogenesis, which are primarily focused on the inhibition of active angiogenesis to starve cancerous cells, target the vascular endothelial growth factor signaling pathway. This aggravates hypoxia within the tumor core and ultimately leads to increased tumor proliferation and metastasis. To overcome this limitation, we developed nanoparticles with antiseptic activity that target tumor vascular abnormalities. METHODS Ferritin-based protein C nanoparticles (PCNs), known as TFG and TFMG, were generated and tested in Lewis lung carcinoma (LLC) allograft and MMTV-PyMT spontaneous breast cancer models. Immunohistochemical analysis was performed on tumor samples to evaluate the tumor vasculature. Western blot and permeability assays were used to explore the role and mechanism of the antitumor effects of PCNs in vivo. For knocking down proteins of interest, endothelial cells were transfected with siRNAs. Statistical analysis was performed using one-way ANOVA followed by post hoc Dunnett's multiple comparison test. RESULTS PCNs significantly inhibited hypoxia and increased pericyte coverage, leading to the inhibition of tumor growth and metastasis, while increasing survival in LLC allograft and MMTV-PyMT spontaneous breast cancer models. The coadministration of cisplatin with PCNs induced a synergistic suppression of tumor growth by improving drug delivery as evidenced by increased blood prefusion and decreased vascular permeability. Moreover, PCNs altered the immune cell profiles within the tumor by increasing cytotoxic T cells and M1-like macrophages with antitumor activity. PCNs induced PAR-1/PAR-3 heterodimerization through EPCR occupation and PAR-1 activation, which resulted in Gα13-RhoA-mediated-Tie2 activation and stabilized vascular tight junctions via the Akt-FoxO3a signaling pathway. CONCLUSIONS Cancer treatment targeting the tumor vasculature by inducing antitumor immune responses and enhancing the delivery of a chemotherapeutic agent with PCNs resulted in tumor regression and may provide an effective therapeutic strategy.
Collapse
Affiliation(s)
- Young Sun Choi
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Daegu, Republic of Korea.,Department of Molecular Pathophysiology, Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.,Nano-Bio Application Team, National Nanofab Center (NNFC), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hyeonha Jang
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Daegu, Republic of Korea.,Department of Molecular Pathophysiology, Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Biki Gupta
- Research Institute of Pharmaceutical Sciences, Kyungpook National Univ., Daegu, Republic of Korea.,Present address Department of Pathology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
| | - Ji-Hak Jeong
- Department of Molecular Pathophysiology, Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.,Research Institute of Pharmaceutical Sciences, Kyungpook National Univ., Daegu, Republic of Korea
| | - Yun Ge
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Daegu, Republic of Korea.,Research Institute of Pharmaceutical Sciences, Kyungpook National Univ., Daegu, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Jong-Sup Bae
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Daegu, Republic of Korea.,Research Institute of Pharmaceutical Sciences, Kyungpook National Univ., Daegu, Republic of Korea
| | - Im-Sook Song
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Daegu, Republic of Korea.,Department of Molecular Pathophysiology, Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.,Research Institute of Pharmaceutical Sciences, Kyungpook National Univ., Daegu, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - You Mie Lee
- BK21 Plus KNU Multi-Omics Creative Drug Research Team, Daegu, Republic of Korea. .,Department of Molecular Pathophysiology, Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea. .,Research Institute of Pharmaceutical Sciences, Kyungpook National Univ., Daegu, Republic of Korea.
| |
Collapse
|
246
|
Macpherson AM, Barry SC, Ricciardelli C, Oehler MK. Epithelial Ovarian Cancer and the Immune System: Biology, Interactions, Challenges and Potential Advances for Immunotherapy. J Clin Med 2020; 9:E2967. [PMID: 32937961 PMCID: PMC7564553 DOI: 10.3390/jcm9092967] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Recent advances in the understanding of immune function and the interactions with tumour cells have led to the development of various cancer immunotherapies and strategies for specific cancer types. However, despite some stunning successes with some malignancies such as melanomas and lung cancer, most patients receive little or no benefit from immunotherapy, which has been attributed to the tumour microenvironment and immune evasion. Although the US Food and Drug Administration have approved immunotherapies for some cancers, to date, only the anti-angiogenic antibody bevacizumab is approved for the treatment of epithelial ovarian cancer. Immunotherapeutic strategies for ovarian cancer are still under development and being tested in numerous clinical trials. A detailed understanding of the interactions between cancer and the immune system is vital for optimisation of immunotherapies either alone or when combined with chemotherapy and other therapies. This article, in two main parts, provides an overview of: (1) components of the normal immune system and current knowledge regarding tumour immunology, biology and their interactions; (2) strategies, and targets, together with challenges and potential innovative approaches for cancer immunotherapy, with attention given to epithelial ovarian cancer.
Collapse
Affiliation(s)
- Anne M. Macpherson
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Simon C. Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia;
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Martin K. Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia
| |
Collapse
|
247
|
Yan Z, Yao ZH, Yao SN, Wang HY, Chu JF, Song M, Zhao S, Liu YY. Camrelizumab plus apatinib successfully treated a patient with advanced esophageal squamous cell carcinoma. Immunotherapy 2020; 12:1161-1166. [PMID: 32814482 DOI: 10.2217/imt-2020-0197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Advanced esophageal squamous cell carcinoma (ESCC) is a lethal disease with poor response to conventional chemotherapy. Immunotherapy showed better activity than chemotherapy in late-line treatment. However, the rate and duration of response are far from satisfactory. The efficacy of an anti-angiogenic agent combined with immunotherapy for ESCC is unknown. Results: A patient with ESCC experienced disease relapse after chemo-radiotherapy. The disease progressed after combined chemotherapy. A combination regimen of the PD-1 inhibitor camrelizumab and the anti-angiogenic agent apatinib was administered. The patient achieved a PET/CT-confirmed durable complete response with mild toxicity. Conclusion: The PD-1 inhibitor combined with the anti-angiogenic agent is effective and safe for the treatment of ESCC. This regimen is worth investigation in clinical trials.
Collapse
Affiliation(s)
- Zheng Yan
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Zhi-Hua Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Shu-Na Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Hai-Ying Wang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Jun-Feng Chu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Ming Song
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Shuang Zhao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Yan-Yan Liu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| |
Collapse
|
248
|
Nagl L, Horvath L, Pircher A, Wolf D. Tumor Endothelial Cells (TECs) as Potential Immune Directors of the Tumor Microenvironment - New Findings and Future Perspectives. Front Cell Dev Biol 2020; 8:766. [PMID: 32974337 PMCID: PMC7466447 DOI: 10.3389/fcell.2020.00766] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/21/2020] [Indexed: 12/30/2022] Open
Abstract
The tumor microenvironment (TME) plays a central role in cancer development and progression. It represents a complex network of cancer cell (sub-)clones and a variety of stromal cell types. Recently, new technology platforms shed light on the cellular composition of the TME at very high resolution and identified a complex landscape of multi-lineage immune cells (e.g., T and B lymphocytes, myeloid cells, and dendritic cells), cancer associated fibroblasts (CAF) and tumor endothelial cells (TECs). A growing body of evidence suggests that metabolically, genetically and on their transcriptomic profile TECs exhibit unique phenotypic and functional characteristics when compared to normal endothelial cells (NECs). Furthermore, the functional role of TECs is multifaceted as they are not only relevant for promoting tumor angiogenesis but have also evolved as key mediators of immune regulation in the TME. Regulatory mechanisms are complex and profoundly impact peripheral immune cell trafficking into the tumor compartment by acting as major gatekeepers of cellular transmigration. Moreover, TECs are associated with T cell priming, activation and proliferation by acting as antigen-presenting cells themselves. TECs are also essential for the formation of tertiary lymphoid structures (TLS) within the tumor, which have recently been associated with treatment response to checkpoint antibody therapy. Further essential characteristics of TECs compared to NECs are their high proliferative potential as well as greatly altered gene expression profile (e.g., upregulation of pro-angiogenic, extracellular matrix remodeling, and stemness genes), which results in enhanced secretion of immunomodulatory cytokines and altered cell-surface receptors [e.g., major histocompatibility complex (MHC) and immune checkpoints]. The TEC phenotype may be rooted in an aggressive tumor micro-milieu based on cellular stress via hypoxia and reactive oxygen species (ROS). Vice versa TECs might modulate TME immunogenicity thereby fostering cancer-associated immune suppression. This review aims to elucidate the currently emergent pathophysiological aspects of TECs with a particular focus on their potential role as regulators of immune cell function in the TME. It is a main future challenge to deeply characterize the phenotypic and functional profile of TECs to illuminate their complex role within the TME. The ultimate goal is the identification of TEC-specific drug targets to improve cancer (immuno-)therapy.
Collapse
Affiliation(s)
- Laurenz Nagl
- Department of Internal Medicine V (Haematology and Oncology), Medical University of Innsbruck, Innsbruck, Austria
| | - Lena Horvath
- Department of Internal Medicine V (Haematology and Oncology), Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Pircher
- Department of Internal Medicine V (Haematology and Oncology), Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Department of Internal Medicine V (Haematology and Oncology), Medical University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute (TKFI), Innsbruck, Austria.,Department of Oncology, Hematology, Rheumatology and Immunoncology, University Hospital Bonn (UKB), Bonn, Germany
| |
Collapse
|
249
|
Li Y, Liu Y, Du B, Cheng G. Reshaping Tumor Blood Vessels to Enhance Drug Penetration with a Multistrategy Synergistic Nanosystem. Mol Pharm 2020; 17:3151-3164. [PMID: 32787273 DOI: 10.1021/acs.molpharmaceut.0c00077] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ying Li
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Ying Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Du
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Genyang Cheng
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
250
|
Trebeschi S, Drago SG, Birkbak NJ, Kurilova I, Cǎlin AM, Delli Pizzi A, Lalezari F, Lambregts DMJ, Rohaan MW, Parmar C, Rozeman EA, Hartemink KJ, Swanton C, Haanen JBAG, Blank CU, Smit EF, Beets-Tan RGH, Aerts HJWL. Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Ann Oncol 2020; 30:998-1004. [PMID: 30895304 PMCID: PMC6594459 DOI: 10.1093/annonc/mdz108] [Citation(s) in RCA: 351] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Immunotherapy is regarded as one of the major breakthroughs in cancer treatment. Despite its success, only a subset of patients responds-urging the quest for predictive biomarkers. We hypothesize that artificial intelligence (AI) algorithms can automatically quantify radiographic characteristics that are related to and may therefore act as noninvasive radiomic biomarkers for immunotherapy response. PATIENTS AND METHODS In this study, we analyzed 1055 primary and metastatic lesions from 203 patients with advanced melanoma and non-small-cell lung cancer (NSCLC) undergoing anti-PD1 therapy. We carried out an AI-based characterization of each lesion on the pretreatment contrast-enhanced CT imaging data to develop and validate a noninvasive machine learning biomarker capable of distinguishing between immunotherapy responding and nonresponding. To define the biological basis of the radiographic biomarker, we carried out gene set enrichment analysis in an independent dataset of 262 NSCLC patients. RESULTS The biomarker reached significant performance on NSCLC lesions (up to 0.83 AUC, P < 0.001) and borderline significant for melanoma lymph nodes (0.64 AUC, P = 0.05). Combining these lesion-wide predictions on a patient level, immunotherapy response could be predicted with an AUC of up to 0.76 for both cancer types (P < 0.001), resulting in a 1-year survival difference of 24% (P = 0.02). We found highly significant associations with pathways involved in mitosis, indicating a relationship between increased proliferative potential and preferential response to immunotherapy. CONCLUSIONS These results indicate that radiographic characteristics of lesions on standard-of-care imaging may function as noninvasive biomarkers for response to immunotherapy, and may show utility for improved patient stratification in both neoadjuvant and palliative settings.
Collapse
Affiliation(s)
- S Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam; GROW School of Oncology and Developmental Biology, Maastricht, The Netherlands; Departments of Radiation Oncology; Radiology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - S G Drago
- Department of Radiology, Netherlands Cancer Institute, Amsterdam; Department of Radiology, Milano-Bicocca University, San Gerardo Hospital, Monza, Italy
| | - N J Birkbak
- The Francis Crick Institute, London; University College London, London, UK; Department of Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - I Kurilova
- Department of Radiology, Netherlands Cancer Institute, Amsterdam; GROW School of Oncology and Developmental Biology, Maastricht, The Netherlands
| | - A M Cǎlin
- Department of Radiology, Netherlands Cancer Institute, Amsterdam; Affidea Romania, Cluj-Napoca, Romania
| | - A Delli Pizzi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam; ITAB Institute for Advanced Biomedical Technologies, University G. d'Annunzio, Chieti, Italy
| | - F Lalezari
- Department of Radiology, Netherlands Cancer Institute, Amsterdam
| | - D M J Lambregts
- Department of Radiology, Netherlands Cancer Institute, Amsterdam
| | | | - C Parmar
- Departments of Radiation Oncology; Radiology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | | | | | - C Swanton
- The Francis Crick Institute, London; University College London, London, UK
| | | | | | - E F Smit
- Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam; GROW School of Oncology and Developmental Biology, Maastricht, The Netherlands
| | - H J W L Aerts
- Department of Radiology, Netherlands Cancer Institute, Amsterdam; Departments of Radiation Oncology; Radiology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.
| |
Collapse
|